CN114401732A - CD38 binding agents and uses thereof - Google Patents

CD38 binding agents and uses thereof Download PDF

Info

Publication number
CN114401732A
CN114401732A CN202080061542.3A CN202080061542A CN114401732A CN 114401732 A CN114401732 A CN 114401732A CN 202080061542 A CN202080061542 A CN 202080061542A CN 114401732 A CN114401732 A CN 114401732A
Authority
CN
China
Prior art keywords
xaa
amino acid
optionally substituted
residue
independently
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202080061542.3A
Other languages
Chinese (zh)
Inventor
L·拉斯泰利
M·E·韦尔施
A·布宁
A·M·K·罗西
T·蓓尔巴索娃
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cleo Pharmaceutical Co ltd
Original Assignee
Cleo Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cleo Pharmaceutical Co ltd filed Critical Cleo Pharmaceutical Co ltd
Publication of CN114401732A publication Critical patent/CN114401732A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/16Blood plasma; Blood serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/64Cyclic peptides containing only normal peptide links

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Cell Biology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Biotechnology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Molecular Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present disclosure provides, inter alia, compounds comprising an antibody binding moiety and a CD38 binding targeting moiety. In some embodiments, provided compounds recruit different types of antibodies to diseased cells, such as cancer cells, and induce immune activity to kill such cells. The provided technology is applicable to the treatment of various diseases including cancer.

Description

CD38 binding agents and uses thereof
Cross Reference to Related Applications
This application claims priority from 62/870,633 No. 62/870,633 filed on 7/03/2019 and U.S. provisional application No. 62/951,765 filed on 12/20/2019, each of which is incorporated herein by reference in its entirety.
Technical Field
The present disclosure provides, inter alia, techniques (e.g., compounds, compositions, and methods thereof) useful, for example, in the treatment of various conditions, disorders, or diseases.
Background
Immune system activity can be utilized to prevent or treat a variety of conditions, disorders and diseases.
Disclosure of Invention
In some embodiments, the present disclosure provides techniques, e.g., compounds, compositions, methods, etc., that are particularly useful for recruiting antibodies to damaged or defective tissues (e.g., tumors, certain wounds, etc.), foreign objects or entities (e.g., infectious agents), etc., including CD38 or fragments thereof. In some embodiments, the provided techniques can trigger, generate, promote, and/or enhance immune system activity, e.g., antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), etc., against target cells, tissues, subjects, and/or entities expressing CD 38. In some embodiments, the present disclosure is directed to the design, preparation, and use of molecules capable of selectively redirecting endogenous antibodies to diseased cells expressing CD38 (e.g., cancer cells) and inducing immune system activity (e.g., antibody-directed cell-mediated immune responses, e.g., cytotoxicity, ADCP, etc.).
In some embodiments, the present disclosure provides an Antibody Recruiting Molecule (ARM) comprising an antibody binding portion, a target binding portion (e.g., a target binding portion that binds to CD 38), and an optional linker portion. In some embodiments, the target binding moiety confers specificity to ARM for its target (e.g., a diseased cell of interest) via, for example, binding to an entity that distinguishes the target from non-targets (e.g., diseased cells from other cell types). In addition, ARM can achieve target-specific recruitment of antibodies (e.g., endogenous antibodies, administered antibodies, etc.) via ABT, and/or trigger, generate, promote, and/or enhance immune activity, such as immune-mediated killing of target cells. In some embodiments, the provided technology includes an ARM comprising a target binding moiety that binds CD38, and can selectively recruit antibodies to a target expressing CD38 (e.g., a cancer cell) and/or trigger, generate, promote, and/or enhance immune activity (e.g., ADCC, ADCP, etc.) against such target cells. In some embodiments, the target cell expressing CD is a cancer cell. In some embodiments, the agents provided herein, such as ARM that binds CD38, are particularly useful for the prevention and/or treatment of conditions, disorders, or diseases associated with CD38, such as various types of cancers associated with CD 38.
Further, the present disclosure encompasses the following recognition: without wishing to be bound by any particular theory, certain immunotherapies that target targets expressing CD38 (e.g., CD38 antibodies) have one or more side effects (e.g., toxicity) due to the reduction and/or depletion of normal cells expressing CD 38. For example, as described herein, in some embodiments, a CD38 antibody, such as Daratumumab (Daratumumab), induces a reduction or depletion of immune effector cells expressing CD 38. Furthermore, the present disclosure demonstrates that the provided technology can recruit immune components and activity to CD38 expressing targets (e.g., cancer cells) to a lesser extent or without significantly reducing or depleting CD38 expressing immune effector cells as compared to CD38 antibodies such as daratumab.
In some embodiments, provided compounds (e.g., ARM) include an antibody binding moiety (universal antibody binding terminus, uABT) that can bind to antibodies of various specificities. Furthermore, such ARM can circumvent the dependence of a particular antibody population and/or adverse effects associated with individual variation of a particular antibody population. In some embodiments, the uABT can bind to an antibodyAn Fc region, and thereby can recruit antibodies with various antigen specificities, among others. In some embodiments, ABT (e.g., uABT) binds to F of IgG CConserved positions present in the regions. In some embodiments, uABT is capable of recruiting all IgG subclasses (IgG1, IgG2, IgG3, IgG 4). In some embodiments, uABT is capable of preferentially recruiting IgG1, IgG2, and/or IgG 4. In some embodiments, the uABT binds to IgG molecules and does not bind to human IgA or IgM. In some embodiments, recruitment of antibodies (e.g., IgG subclasses) is dependent on the administered dose of ARM, and/or is independent of the level of antibodies with specific Fab regions in the individual. In some embodiments, suitable ABTs are those described in WO 2019/023501, the antibody binding portions of which (e.g., various ABTs, including uABT) are incorporated herein by reference. One of skill in the art will appreciate that a variety of antibody binding moieties are available and can be utilized in accordance with the present disclosure.
In addition, the ARM can recruit an antibody, and the recruited antibody provides one or more immune activities, e.g., via one or more antibody-mediated immune mechanisms. In some embodiments, the recruited antibody recruits and/or interacts with and/or activates an Fc receptor of an immune cell. In some embodiments, the recruited antibodies recruit and activate immune cells and inhibit and/or target diseased cells, such as cancer cells. In some embodiments, the provided agent (e.g., ARM) induces antibody-dependent effector function. In some embodiments, the provided agents (e.g., ARM) induce Complement Dependent Cytotoxicity (CDC). In some embodiments, the provided agents (e.g., ARM) induce direct cytotoxicity. In some embodiments, the provided agent (e.g., ARM) inhibits a biological function associated with steric blockade. In some embodiments, the provided agents (e.g., ARM) induce antibody-dependent cell-mediated viral inhibition (ADCVI). In some embodiments, the provided agent (e.g., ARM) induces ADCC and kills cancer cells. In some embodiments, the provided agents (e.g., ARM) induce ADCP and kill cancer cells. In some embodiments, the provided agents (e.g., ARM) induce both ADCC and ADCP.
In some embodiments, the present disclosure provides a medicament comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
optionally a linker moiety.
In some embodiments, the target binding moiety may bind CD 38. In some embodiments, the antibody binding portion can bind to two or more antibodies having different Fab regions. In some embodiments, an antibody binding moiety can bind to two or more antibodies having different antigen specificities. In some embodiments, the antibody binding portion can bind to the Fc region of various antibodies. In some embodiments, an antibody binding moiety (e.g., a universal antibody binding moiety) binds to the Fc region of an antibody. In some embodiments, an antibody binding moiety (e.g., a universal antibody binding moiety) binds to a conserved Fc region of an antibody. In some embodiments, the antibody binding moiety binds to the Fc region of an IgG antibody. In some embodiments, the antibody may still perform all or substantially all or most of its biological functions after binding to the antibody binding moiety (e.g., binding at the Fc region). For example, an antibody upon binding to an antibody binding moiety may recruit and/or activate immune cells, e.g., via interaction with various Fc receptors.
In some embodiments, the present disclosure provides compounds having general formula I:
Figure BDA0003526174380000031
or a pharmaceutically acceptable salt thereof, wherein the variables are independently as defined and described herein. In some embodiments, the provided agent is a compound of formula I or a salt thereof.
In some embodiments, provided agents (e.g., compounds of formula I) are those having
Figure BDA0003526174380000032
A compound of structure (la), or a pharmaceutically acceptable salt thereof, wherein the variables are as defined and described in the disclosure.
In some embodiments, provided agents are compounds of formula I-a or salts thereof. In some embodiments, the present disclosure provides a compound of formula I-a:
Figure BDA0003526174380000041
or a pharmaceutically acceptable salt thereof, wherein the variables are as defined and described in the disclosure. In some embodiments, provided compounds of formula I are compounds of formula I-a.
In some embodiments, provided agents are compounds of formula I-b or salts thereof. In some embodiments, the present disclosure provides a compound of formula I-b:
Figure BDA0003526174380000042
or a pharmaceutically acceptable salt thereof, wherein the variables are as defined and described in the disclosure. In some embodiments, provided compounds of formula I are compounds of formula I-b.
In some embodiments, the agents and compounds provided by the present disclosure and pharmaceutically acceptable compositions thereof are effective for recruiting antibodies to diseased cells, such as cancer cells. In some embodiments, the present disclosure provides compounds having general formula II:
Figure BDA0003526174380000043
Or a pharmaceutically acceptable salt thereof, wherein the variables are as defined and described herein. In some embodiments, the provided agent is a compound of formula II or a salt thereof. In some embodiments, a provided compound of formula I is a provided compound of formula II or a salt thereof. In some embodiments, the compound having the structure of formula I-a is a compound of formula II.
In some embodiments, the present disclosure provides compounds having general formula III:
Figure BDA0003526174380000051
or a pharmaceutically acceptable salt thereof, wherein the variables are as defined and described herein. In some embodiments, the provided agent is a compound of formula III or a salt thereof. In some embodiments, a provided compound of formula I is a provided compound of formula III, or a salt thereof. In some embodiments, the compound having the structure of formula I-b is a compound of formula III.
The compounds of the present disclosure and pharmaceutically acceptable compositions thereof are useful in the treatment of a variety of diseases, disorders, or conditions. Such diseases, disorders, or conditions include those described herein. In some embodiments, the condition, disorder or disease is cancer.
Drawings
FIG. 1 the compounds provided recruit antibodies to target cells.
Figure 2 provided compounds can recruit antibodies to target cells and activate effector cells.
FIG. 3 provides compounds that kill target cells.
Figure 4. the provided compounds did not significantly deplete effector cells. Darunavir single antibody: 3 μ g/mL to 0.1 μ g/mL. I-9: 300nM to 10 nM. IvIG: 10. mu.g/mL.
Figure 5. the technique provided has low toxicity. A. Frequency of dead NK cells. B. Dead NK cell frequency normalized to DMSO-treated control. The CD38 ARM is I-17.
FIG. 6. the technique provided is effective in killing cancer cells. Frequency of dead SUDHL-4 cells in nk-SUDHL-4 co-cultures. B. SuDHL-4 cell death frequency in NK-SUDHL-4 co-cultures normalized to DMSO-treated controls. The CD38 ARM is I-17.
FIG. 7. techniques are provided to reduce the number of plasma cells. The CD38 ARM is I-17.
Figure 8. the provided technology has no or low levels of undesired NK cell suicide. The CD38 ARM is I-17.
FIG. 9. the technique provided is effective in reducing the number of target cells. Without intending to be limited by theory, the count of Daudi cells expressing CD38 in the intraperitoneal cavity of SCID mice served as a readout for macrophage-mediated I-17 dependent phagocytosis. From left to right: comparison; IVIG 10 mg/mouse sub-Q; i-171 mg/kg + IVIG (10 mg/mouse sub-Q); i-1710 mg/kg + IVIG (10 mg/mouse sub-Q); i-1730 mg/kg + IVIG (10 mg/mouse sub-Q); daratumab. ***: p is less than 0.001.
FIG. 10 Activity of CIML NK cells frozen in combination with I-17 and restored against MOLP-8 cells. From left to right: untreated; darunavir monoclonal antibody; cryopreserved CIML NK I-17 added to the assay; and CIML NK cryopreserved with I-17.
Detailed Description
1. General description of certain embodiments
In some embodiments, the present disclosure provides agents, such as ARM, that include a target binding moiety that can bind to CD 38. In some embodiments, provided agents (e.g., ARM) include a universal antibody binding moiety that can bind to antibodies with different Fab structures. In some embodiments, the disclosure provides agents (e.g., ARM) that include an antibody binding portion that binds to an antibody (e.g., an Fc region of an antibody), and such binding of the antibody does not interfere with one or more immune activities of the antibody, e.g., interaction with an Fc receptor (e.g., CD16a), recruitment of effector cells (e.g., NK cells (e.g., for ADCC), macrophages (e.g., for ADCP)), etc. As will be appreciated by those of skill in the art, the techniques (agents, compounds, compositions, methods, etc.) provided by the present disclosure may provide various advantages, for example, the provided techniques may utilize antibodies with various Fab regions in the immune system to avoid or minimize adverse effects of antibody changes between patient populations, may trigger and/or enhance immune activity against a target (e.g., kill a target diseased cell, such as a cancer cell), and/or have low toxicity (e.g., low complement activation, significantly less normal cells expressing CD38 (e.g., effector cells) reduction) compared to certain antibody therapeutics.
In some embodiments, the provided techniques are suitable for modulating immune activity, such as ADCC, ADCP and combinations thereof, against a target (diseased cells, foreign subject or entity, etc.) comprising CD 38. In some embodiments, the techniques of the present disclosure are suitable for recruiting antibodies to cancer cells, particularly cancer cells expressing CD 38. In some embodiments, the provided techniques are suitable for modulating ADCC against a target cell (e.g., a diseased cell, such as a cancer cell). In some embodiments, the provided techniques are suitable for modulating ADCP against a target cell (e.g., a diseased cell, such as a cancer cell). In some embodiments, provided agents can inhibit protein activity. In some embodiments, the target binding moiety is an inhibitor moiety. In some embodiments, the target binding moiety is an enzyme inhibitor moiety.
In some embodiments, the present disclosure provides a medicament comprising:
(ii) an antibody-binding moiety,
a target binding moiety that binds to CD38, and
the optional presence of a linker moiety or moieties,
wherein the antibody binding portion can bind to two or more antibodies having different Fab regions.
In some embodiments, the present disclosure provides a medicament comprising:
(ii) an antibody-binding moiety,
a target binding moiety that binds to CD38, and
the optional presence of a linker moiety or moieties,
wherein the antibody binding portion can bind to two or more antibodies having different Fab regions.
In some embodiments, provided agents include two or more antibody binding moieties. In some embodiments, provided agents include two or more target binding moieties.
The antibody binding portion may interact with any portion of the antibody. In some embodiments, the antibody binding moiety binds to the Fc region of an antibody. In some embodiments, the antibody binding portion binds to a conserved Fc region of the antibody. In some embodiments, the antibody binding moiety binds to the Fc region of an IgG antibody. As will be appreciated by those skilled in the art, a variety of antibody binding moieties, linkers, and target binding moieties can be utilized in accordance with the present disclosure. Furthermore, as shown in the examples, in some embodiments, the present disclosure provides antibody binding moieties, linkers, and target binding moieties, and combinations thereof, that are particularly useful and effective for constructing ARM molecules to recruit antibodies to target cells and/or to trigger, generate, promote, and/or enhance immune system activity against target cells (e.g., diseased cells, e.g., cancer cells).
In some embodiments, the present disclosure provides antibody binding portions that can bind to an Fc region that binds to an Fc receptor (e.g., fcyriiia, CD16a, etc.) and/or agents that include the antibody binding portions (e.g., compounds of the various formulae described in the present disclosure, ARM molecules of the present disclosure, etc.). In some embodiments, an agent that binds to a moiety that comprises a complex of an Fc region and an Fc receptor and/or comprises the antibody binding moiety is provided. In some embodiments, the present disclosure provides a composite comprising:
a medicament, comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
the optional presence of a linker moiety or moieties,
an Fc region, and
an Fc receptor.
In some embodiments, the antibody binding portion can bind to CD38, and/or the antibody binding portion of the agent can bind to two or more antibodies having different Fab regions.
In some embodiments, the Fc region is an Fc region of an endogenous antibody of the subject. In some embodiments, the Fc region is that of an exogenous antibody. In some embodiments, the Fc region is that of the administered agent. In some embodiments, the Fc receptor belongs to a diseased cell in the subject. In some embodiments, the Fc receptor belongs to a cancer cell of the subject.
In certain embodiments, the present disclosure provides a compound of formula I:
Figure BDA0003526174380000081
or a pharmaceutically acceptable salt thereof, wherein:
each of a and b is independently 1 to 200;
each ABT is independently an antibody binding moiety;
l is a bivalent or multivalent linker moiety linking ABT to TBT; and is
Each TBT is independently a target binding moiety.
In some embodiments, the ABT is a universal antibody binding moiety.
In some embodiments, the antibody binding portion comprises one or more amino acid residues. In some embodiments, the antibody-binding moiety is or includes a peptide moiety. In some embodiments, the antibody-binding moiety is or includes a cyclic peptide moiety. In some embodiments, such antibody binding moieties include one or more natural amino acid residues. In some embodiments, such antibody binding moieties include one or more non-natural, natural amino acid residues.
In some embodiments, the amino acid has the structure of formula a-I:
NH(Ra1)-La1-C(Ra2)(Ra3)-La2-COOH,
A-I
or a salt thereof, wherein:
Ra1、Ra2、Ra3each of which is independently-La-R′;
La1And La2Each of which is independently La
Each LaIndependently a covalent bond, or is selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R') 2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from: c3-20A cycloaliphatic ring; c6-20An aryl ring; a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 20 membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, the amino acid analogs are compounds in which the amino group and/or the carboxylic acid group are independently replaced with an optionally substituted aliphatic or heteroaliphatic moiety. As will be appreciated by those of skill in the art, many amino acid analogs that mimic the structure, properties, and/or function of an amino acid are described in the art and can be utilized in accordance with the present disclosure.
In some embodiments, the antibody-binding moiety is a cyclic peptide moiety. In some embodiments, the present disclosure provides a compound of formula I-a:
Figure BDA0003526174380000091
or a salt thereof, wherein:
each Xaa is independently a residue of an amino acid or amino acid analog;
t is 0 to 50;
z is 1 to 50;
l is a linker moiety;
TBT is a target binding moiety;
each RcIndependently is-La-R′;
Each of a and b is independently 1 to 200;
each LaIndependently a covalent bond, or is selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R') 2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from: c3-20A cycloaliphatic ring; c6-20An aryl ring; a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 20 membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, a is 1. In some embodiments, b is 1. In some embodiments, a is 1 and b is 1, and the compounds of formula I-a have the following structure:
Figure BDA0003526174380000101
in some embodiments, each residue (e.g., Xaa) is independently a residue of an amino acid or an amino acid analog, wherein the amino acid or the amino acid analog has H-La1-La1-C(Ra2)(Ra3)-La2-La2-H structure or a salt thereof. In some embodiments, the amino acid has NH (R)a1)-La1-C(Ra2)(Ra3)-La2-COOH structure orA salt thereof. In some embodiments, the amino acid analog has H-La1-La1-C(Ra2)(Ra3)-La2-La2-H structure or a salt thereof. In some embodiments, in such amino acid analogs, the first-La1- (bonded to-H in said formula) other than-N (R)a1) - (e.g. being optionally substituted divalent C)1-6Aliphatic group). In some embodiments, in H-La1-La1-in, -La1-La1-is bonded to-H via an atom other than nitrogen. In some embodiments, at-La2-La2in-H, -La2-La2-is not bonded to-H via-C (O) O-. In some embodiments, each residue (e.g., each Xaa in formula I-a) is independently a residue of an amino acid having the structure of formula A-I.
In some embodiments, each Xaa independently has-La1-La1-C(Ra2)(Ra3)-La2-La2-a structure. In some embodiments, each Xaa independently has-LaX1-La1-C(Ra2)(Ra3)-La2-LaX2-structure wherein LaX1Is optionally substituted-NH-, optionally substituted-CH2-、-N(Ra1) -or-S-, LaX2Is optionally substituted-NH-, optionally substituted-CH2-、-N(Ra1) -or-S-, and each other variable is independently as described herein. In some embodiments, LaX1Is optionally substituted-NH-or-N (R)a1) -. In some embodiments, LaX1Is optionally substituted-CH2-or-S-. In some embodiments, LaX2Is optionally substituted-NH-, optionally substituted-CH2-、-N(Ra1) -or-S-. In some embodiments, optionally substituted-CH2is-C (O) -. In some embodiments, optionally substituted-CH2-is not-C (O) -. In some embodiments, LaX2is-C (O) -. In some embodiments, each Xaa independently has-N (R)a1)-La1-C(Ra2)(Ra3)-La2-CO-structure.
In many embodimentsTwo or more residues (e.g., two or more Xaa residues) are linked together to form one or more cyclic structures. For example, various compounds in table 1 include attached residues. The residue may optionally be linked via a linker (e.g., L) at any suitable positionT) And (4) connecting. For example, a bond between two residues may link each residue independently at its N-terminus, C-terminus, a point on the backbone, or a point on the side chain, etc. In some embodiments, for example, two or more side chains of a residue in a compound of formula I-a (e.g., R of one amino acid residue) a2Or Ra3With another amino acid residuea2Or Ra3) Optionally together forming a bridge (e.g., in the various compounds in table 1, etc.), for example, in some embodiments, two cysteine residues form an-S-bridge as is commonly observed in native proteins. In some embodiments, the formed bridge has LbStructure of, wherein LbIs L as described in this disclosurea. In some embodiments, LbEach end of (a) is independently attached to a backbone atom of a cyclic peptide (e.g., - (Xaa) in formula I-azThe ring atoms of the ring formed). In some embodiments, LbIncluding the R group (e.g. when LbIs represented by the formula (II) or (III)2-or-N (R) -when substituted), wherein the R group is substituted with an R group attached to the backbone atom (e.g. R when R is R)a1、Ra2、Ra3Etc.) and their intermediate atoms together form a ring. In some embodiments, LbTo the ring via the side chain of an amino acid residue (e.g. Xaa in formula I-a), e.g. by- (Xaa) in formula I-az-the ring formed. In some embodiments, such side chains include amino groups or carboxylic acid groups. In some embodiments, LTIs L as described hereinb. In some embodiments, a key (e.g., L)bOr LT) Linking the side chain of the residue to the N-terminus or C-terminus. In some embodiments, the side chain linking the residue to an amino group. In some embodiments, the side chain linking the residue to the α -amino group. In some embodiments, as illustrated herein, a key (e.g., L) bOr LT) is-CH2-C (O) -. At one endIn some embodiments, -CH2-S-bonded to a side chain, e.g. to a cysteine residue, and-c (o) -bonded to an amino group, e.g. an alpha-amino group of a residue. In some embodiments, a key (e.g., L)bOr LT) Is optionally substituted-CH2-S-CH2-C (O) -NH-, wherein each end is bonded to the α -carbon of the residue. In some embodiments, -NH-belongs to the alpha-amino group of a residue (e.g., the N-terminal residue).
In some embodiments of the present invention, the,
Figure BDA0003526174380000121
is an antibody-binding moiety: (
Figure BDA0003526174380000122
Binding to an antibody). In some embodiments of the present invention, the,
Figure BDA0003526174380000123
is a universal antibody binding moiety. In some embodiments of the present invention, the,
Figure BDA0003526174380000124
is a universal antibody binding moiety that can bind to antibodies having different Fab regions. In some embodiments of the present invention, the,
Figure BDA0003526174380000125
is a universal antibody binding moiety that can bind to an Fc region. In some embodiments, the antibody-binding moiety, for example, has
Figure BDA0003526174380000126
The universal antibody binding portion of the structure can bind to an Fc region that binds to an Fc receptor. In some embodiments, the antibody-binding moiety, for example, has
Figure BDA0003526174380000127
The antibody-binding portion of the structure has the following structure:
Figure BDA0003526174380000128
in some embodiments of the present invention, the,
Figure BDA0003526174380000129
has the following structure:
Figure BDA00035261743800001210
in certain embodiments, the present disclosure provides a compound of formula II:
Figure BDA0003526174380000131
or a pharmaceutically acceptable salt thereof, wherein:
R1、R3And R5Each of which is independently hydrogen or an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or:
R1and R1' optionally together with its intervening carbon atoms form a 3-to 8-membered optionally substituted saturated or partially unsaturated spirocyclic carbocyclic ring or a 3-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R3and R3' optionally together with its intervening carbon atoms form a 3-to 8-membered optionally substituted saturated or partially unsaturated spirocyclic carbocyclic ring or a 3-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
r bound to the same carbon atom5Group and R5' the group optionally together with its intervening carbon atoms forms a 3-to 8-membered optionally substituted saturated or partially unsaturated spirocyclic carbocyclic ring or has 1 to 2 hetero atoms independently selected from nitrogen, oxygen or sulfur An atomic 3 to 8 membered saturated or partially unsaturated spirocyclic heterocycle; or
Two R5The radicals optionally forming C together with their intermediate atoms1-10An optionally substituted divalent linear or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 3 methylene units of said chain are independently and optionally substituted by-S-, -SS-, -N (R) -, -O-, -C (O) -, -OC (O) -, -C (O) O-, -C (O) N (R) -, -N (R) C (O) -, -S (O)2-or-Cy1-substitution, wherein each-Cy1-independently is a 5-to 6-membered heteroarylene having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R1′、R3' and R5Each of' is independently hydrogen or optionally substituted C1-3An aliphatic group;
R2、R4and R6Each of which is independently hydrogen or optionally substituted C1-4An aliphatic group, or:
R2and R1Optionally together with their intermediate atoms, form a 4-to 8-membered optionally substituted saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R4and R3Optionally together with their intermediate atoms, form a 4-to 8-membered optionally substituted saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or
R6Group and its adjacent R5The groups optionally together with their intermediate atoms form a 4-to 8-membered optionally substituted saturated or partially unsaturated monocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
L1To connect to
Figure BDA0003526174380000141
A trivalent linker moiety of (a);
L2is a covalent bond or C1-30An optionally substituted divalent linear or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 10 methylene units of said chain are independently and optionally substituted by-S-, -N(R)-、-O-、-C(O)-、-OC(O)-、-C(O)O-、-C(O)N(R)-、-N(R)C(O)-、-S(O)-、-S(O)2-、
Figure BDA0003526174380000142
or-Cy1-substitution, wherein each-Cy1-independently is a 5-to 6-membered heteroarylene having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
TBT is a target binding moiety; and is
Each of m and n is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
In some embodiments, the antibody-binding moiety is or includes a peptide moiety. In some embodiments, the present disclosure provides a compound having the structure of formula I-b:
Figure BDA0003526174380000143
or a salt thereof, wherein:
each Xaa is independently a residue of an amino acid or amino acid analog;
each z is independently 1 to 50;
each L is independently a linker moiety;
the TBT is the binding moiety of the target,
each RcIndependently is-La-R′;
Each of a1 and a2 is independently 0 or 1, wherein at least one of a1 and a2 is not 0;
each of a and b is independently 1 to 200;
each LaIndependently a covalent bond, or is selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R') 2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from: c3-20A cycloaliphatic ring; c6-20An aryl ring; a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 20 membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, a1 is 1. In some embodiments, a2 is 1. In some embodiments, b is 1. In some embodiments, the compounds of formula I-b have the following structure:
Figure BDA0003526174380000151
in some embodiments, the compounds of formula I-b have the following structure:
Figure BDA0003526174380000161
in some embodiments, the compounds of formula I-b have the following structure:
Figure BDA0003526174380000162
in some embodiments, the compounds of formula I-b have the following structure:
Figure BDA0003526174380000163
in some embodiments, each residue (e.g., each Xaa in formulas I-a, I-b, etc.) is independently a residue of an amino acid having the structure of formula A-I. In some embodiments, each Xaa independently has-N (R)a1)-La1-C(Ra2)(Ra3)-La2-CO-structure. In some embodiments, for example, two or more side chains of an amino acid residue (e.g., R of an amino acid residue) in a compound of formula I-aa2Or Ra3With another amino acid residuea2Or Ra3) Optionally forming a bridge (e.g., the various compounds in table 1) together, e.g., in some embodiments, two cysteine residues form an-S-bridge as is commonly observed in native proteins. In some embodiments, the formed bridge has L bStructure of, wherein LbIs L as described in this disclosurea. In some embodiments, LbEach end of (a) is independently attached to a backbone atom of a cyclic peptide (e.g., - (Xaa) in formula I-azThe ring atoms of the ring formed). In some embodiments, LbIncluding the R group (e.g. when LbIs represented by the formula (II) or (III)2-or-N (R) -when substituted), wherein the R group is substituted with an R group attached to the backbone atom (e.g. R when R is R)a1、Ra2、Ra3Etc.) and their intermediate atoms together form a ring. In some embodiments, LbVia side chains of amino acid residues (e.g. of formula I)Xaa in-a) is attached to the ring, e.g. by- (Xaa) in formula I-bz-the ring formed. In some embodiments, such side chains include amino groups or carboxylic acid groups.
In some embodiments, Rc- (Xaa) z-is an antibody binding moiety (R)c- (Xaa) z-H binding to an antibody). In some embodiments, Rc- (Xaa) z-is a universal antibody binding moiety. In some embodiments, Rc- (Xaa) z-is a universal antibody binding moiety that can bind to antibodies having different Fab regions. In some embodiments, Rc- (Xaa) z-is a universal antibody binding moiety that can bind to an Fc region. In some embodiments, the antibody binding moiety, e.g., with RcThe generic antibody binding portion of the- (Xaa) z-structure can bind to an Fc region that binds to an Fc receptor. In some embodiments, R c- (Xaa) z-has the structure:
Figure BDA0003526174380000164
in some embodiments, Rc- (Xaa) z-L-has the structure:
Figure BDA0003526174380000165
in certain embodiments, the present disclosure provides a compound of formula III:
Figure BDA0003526174380000171
or a pharmaceutically acceptable salt thereof, wherein:
R7each of which is independently hydrogen or an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or:
r bound to the same carbon atom7Group and R7' the group optionally together with its intervening carbon atoms forms a 3-to 8-membered optionally substituted saturated or partially unsaturated spirocyclic carbocyclic ring or a 3-to 8-membered optionally substituted saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R7each of' is independently hydrogen or optionally substituted C1-3An aliphatic group;
R8Each of which is independently hydrogen or optionally substituted C1-4An aliphatic group, or:
R8group and R adjacent thereto7The groups optionally together with their intermediate atoms form a 4-to 8-membered optionally substituted saturated or partially unsaturated monocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
R9is hydrogen, optionally substituted C1-3Aliphatic radical or-C (O) - (optionally substituted C)1-3Aliphatic groups);
L3to connect to
Figure BDA0003526174380000172
A divalent linker group to TBT;
TBT is a target binding moiety; and is
o is 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
2. Definition of
The compounds of the present disclosure include compounds generally described herein and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For the purposes of this disclosure, chemical elements are identified according to the periodic table of elements, CAS version, Handbook of Chemistry and Physics, 75 th edition. In addition, the general principles of Organic Chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books (University Science Books), sudox (sautalito): 1999 and "March's Advanced Organic Chemistry", 5 th edition, editor: smith, m.b. and March, j., John Wiley parent-child publishing company (John Wiley & Sons), new york: 2001.
As used herein in this disclosure, unless the context clearly dictates otherwise, (i) the terms "a" and (an) "are to be understood as meaning" at least one "; (ii) the term "or" may be understood to mean "and/or"; (iii) the terms "comprising," including, "" containing, "" whether used with "or without limitation," and "containing" whether used with "or without limitation," are understood to encompass the recited components or steps, whether presented alone or with one or more additional components or steps; (iv) the term "another" can be understood to mean at least an additional/second one or more; (v) the terms "about" and "approximately" are understood to allow for a standard deviation as understood by those skilled in the art; and (vi) when ranges are provided, the endpoints are included. Unless otherwise specified, the compounds described herein may be provided and/or utilized in the form of a salt, specifically a pharmaceutically acceptable salt.
Aliphatic group: as used herein, "aliphatic group" means a straight-chain (i.e., unbranched) or branched substituted or unsubstituted hydrocarbon chain that is fully saturated or contains one or more units of unsaturation, or a substituted or unsubstituted monocyclic, bicyclic, or polycyclic hydrocarbon ring that is fully saturated or contains one or more units of unsaturation, but which is not aromatic, or a combination thereof. In some embodiments, the aliphatic group contains 1 to 50 aliphatic carbon atoms. In some embodiments, the aliphatic group contains 1 to 20 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1 to 10 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1 to 9 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1 to 8 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1 to 7 aliphatic carbon atoms. In other embodiments, the aliphatic group contains 1 to 6 aliphatic carbon atoms. In still other embodiments, the aliphatic group contains 1 to 5 aliphatic carbon atoms, and in still other embodiments, the aliphatic group contains 1, 2, 3, or 4 aliphatic carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched substituted or unsubstituted alkyl, alkenyl, alkynyl groups and mixtures thereof, such as (cycloalkyl) alkyl, (cycloalkenyl) alkyl or (cycloalkyl) alkenyl.
Alkenyl: as used herein, the term "alkenyl" refers to an aliphatic group as defined herein having one or more double bonds.
Alkyl groups: as used herein, the term "alkyl" has its ordinary meaning in the art and can include saturated aliphatic groups, including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl groups (alicyclic groups), cycloalkyl groups substituted with alkyl groups, and alkyl groups substituted with cycloalkyl groups. In some embodiments, the alkyl group has 1 to 100 carbon atoms. In certain embodiments, the straight or branched chain alkyl group has about 1 to 20 carbon atoms in its backbone (e.g., straight is C)1-C20Branched chain is C2-C20) And alternatively about 1 to 10. In some embodiments, cycloalkyl rings have about 3 to 10 carbon atoms in their ring structure, wherein such rings are monocyclic, bicyclic, or polycyclic, and alternatively have about 5, 6, or 7 carbon atoms in the ring structure. In some embodiments, the alkyl group can be a lower alkyl group, wherein the lower alkyl group includes 1 to 4 carbon atoms (e.g., straight chain lower alkyl is C)1-C4)。
Alkynyl: as used herein, the term "alkynyl" refers to an aliphatic group as defined herein having one or more triple bonds.
Aryl: as used herein, the term "aryl", alone or as used as part of a larger moiety in "aralkyl", "aralkoxy", or "aryloxyalkyl", refers to a monocyclic, bicyclic, or polycyclic ring system having a total of five to thirty ring members, wherein at least one ring in the system is aromatic. In some embodiments, aryl is a monocyclic, bicyclic, or polycyclic ring system having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, and wherein each ring in the system contains 3 to 7 ring members. In some embodiments, aryl is biaryl. The term "aryl" is used interchangeably with the term "aryl ring". In certain embodiments of the present disclosure, "aryl" refers to aromatic ring systems including, but not limited to, phenyl, biphenyl, naphthyl, binaphthyl, anthracenyl, and the like, which may bear one or more substituents. As used herein, also included within the scope of the term "aryl" are groups in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthalimide, phenanthridinyl, or tetrahydronaphthyl, and the like.
A cycloaliphatic group: the terms "cycloaliphatic", "carbocycle", "carbocyclyl", and "carbocycle" are used interchangeably and, as used herein, refer to a saturated or partially unsaturated, but non-aromatic, cycloaliphatic monocyclic, bicyclic, or polycyclic ring system as described herein, having from 3 to 30 ring members, unless otherwise specified. Cycloaliphatic groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, cycloheptenyl, cyclooctyl, cyclooctenyl, norbornyl, adamantyl, and cyclooctadienyl. In some embodiments, the cycloaliphatic group has from 3 to 6 carbon atoms. In some embodiments, the cycloaliphatic group is saturated and is cycloalkyl. The term "cycloaliphatic" may also encompass an aliphatic ring fused to one or more aromatic or non-aromatic rings (e.g., decahydronaphthyl or tetrahydronaphthyl). In some embodiments, the cycloaliphatic group is bicyclic. In some embodiments, the cycloaliphatic group is tricyclic. In some embodiments, the cycloaliphatic group is polycyclic. In some embodiments, "cycloaliphatic" refers to a fully saturated or non-aromatic C containing one or more units of unsaturation with a single point of attachment to the rest of the molecule 3-C6Monocyclic hydrocarbon or C8-C10Bicyclic or polycyclic hydrocarbons, or C, fully saturated or containing one or more units of unsaturation, but not aromatic, having a single point of attachment to the rest of the molecule9-C16Polycyclic hydrocarbons.
The administration scheme is as follows: as used herein, a "dosing regimen" or "treatment regimen" refers to a group of unit doses (typically more than one) that are administered individually to a subject, typically at spaced apart time periods. In some embodiments, a given therapeutic agent has a recommended dosing regimen, which may involve one or more doses. In some embodiments, the dosing regimen comprises a plurality of doses, each of which is spaced apart from each other for a period of time of the same length; in some embodiments, the dosing regimen comprises a plurality of doses and at least two different time periods spaced apart by an individual dose. In some embodiments, all doses within a dosing regimen are of the same unit dose amount. In some embodiments, different doses within a dosing regimen have different amounts. In some embodiments, the dosing regimen comprises a first dose in an amount of a first dose, followed by one or more additional doses in an amount of a second dose that is different from the amount of the first dose. In some embodiments, the dosing regimen comprises a first dose in the amount of the first dose followed by one or more additional doses in the amount of a second dose that is the same as the amount of the first dose.
Heteroaliphatic group: as used herein, the term "heteroaliphatic" has its ordinary meaning in the art, and refers to an aliphatic group as described herein in which one or more carbon atoms are independently replaced with one or more heteroatoms (e.g., oxygen, nitrogen, sulfur, silicon, phosphorus, etc.). In some embodiments, selected from C, CH2And CH3Independently by one or more heteroatoms (including oxidized and/or substituted forms thereof). In some embodiments, the heteroaliphatic group is a heteroalkyl group. In some embodiments, the heteroaliphatic group is a heteroalkenyl group.
Heteroalkyl group: as used herein, the term "heteroalkyl" has its ordinary meaning in the art and refers to an alkyl group as described herein in which one or more carbon atoms are independently replaced with one or more heteroatoms (e.g., oxygen, nitrogen, sulfur, silicon, phosphorus, etc.). Examples of heteroalkyl groups include, but are not limited to, alkoxy, poly (ethylene glycol) -, amino substituted with alkyl, tetrahydrofuranyl, piperidinyl, morpholinyl, and the like.
Heteroaryl group: as used herein, the terms "heteroaryl" and "heteroar-", used alone or as part of a larger moiety (e.g., "heteroaralkyl" or "heteroaralkoxy"), refer to monocyclic, bicyclic, or polycyclic ring systems having a total of five to thirty ring members, wherein at least one ring in the system is aromatic and at least one aromatic ring atom is a heteroatom. In some embodiments, heteroaryl is a group having 5 to 10 ring atoms (i.e., monocyclic, bicyclic, or polycyclic), in some embodiments 5, 6, 9, or 10 ring atoms. In some embodiments, heteroaryl groups have 6, 10, or 14 pi electrons in common in the ring array; and has one to five heteroatoms in addition to carbon atoms. Heteroaryl groups include, but are not limited to, thienyl, furyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. In some embodiments, heteroaryl is a heterobiaryl, such as bipyridyl and the like. As used herein, the terms "heteroaryl" and "heteroar-" also encompass groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, wherein the linking group or point of attachment is on the heteroaromatic ring. Non-limiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzothiazolyl, quinolinyl, isoquinolinyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolyl, tetrahydroisoquinolyl, and pyrido [2, 3-b ] -1, 4-oxazin-3 (4H) -one. Heteroaryl groups can be monocyclic, bicyclic, or polycyclic. The term "heteroaryl" may be used interchangeably with the terms "heteroaryl ring", "heteroaryl group" or "heteroaromatic", any of which terms comprises an optionally substituted ring. The term "heteroaralkyl" refers to an alkyl group substituted with a heteroaryl group, wherein the alkyl and heteroaryl portions are independently optionally substituted.
Heteroatom: as used herein, the term "heteroatom" means an atom that is not carbon or hydrogen. In some embodiments, the heteroatom is boron, oxygen, sulfur, nitrogen, phosphorus, or silicon (including various forms of such atoms, e.g., oxidized forms (e.g., of nitrogen, sulfur, phosphorus, or silicon), quaternized forms of basic nitrogen, or a heterocyclic ring (e.g., N, as in 3, 4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl), or NR+(e.g., in N-substituted pyrrolidinyl), and the like). In some embodiments, the heteroatom is oxygen, sulfur, or nitrogen.
Heterocyclic ring: as used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic radical" and "heterocyclic ring" are used interchangeably as used herein and refer to a monocyclic, bicyclic or polycyclic ring moiety (e.g., 3 to 30 membered) that is saturated or partially unsaturated and has one or more heteroatom ring atoms. In some embodiments, heterocyclyl is a stable 5-to 7-membered monocyclic or 7-to 10-membered bicyclic heterocyclic moiety that is saturated or partially unsaturated and that has one or more (preferably one to four) heteroatoms as defined above in addition to carbon atoms. The term "nitrogen" when used in conjunction with a ring atom of a heterocyclic ring includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0 to 3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen may be N (as in 3, 4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or +NR (as in N-substituted pyrrolidinyl). The heterocyclic ring may be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure, and any of the ring atoms may be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic groups include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepine, oxazepine, thiazepine, morpholinyl, and quinuclidinyl. The terms "heterocycle (heterocyclic)", "heterocyclyl ring (heterocyclo)ycycling), "heterocyclyl group," "heterocyclic moiety" and "heterocyclic radial" are used interchangeably herein and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl or tetrahydroquinolinyl. The heterocyclic group may be monocyclic, bicyclic or polycyclic. The term "heterocyclylalkyl" refers to an alkyl group substituted with a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
Lower alkyl group: the term "lower alkyl" refers to C1-4Straight-chain or branched-chain alkyl. Examples of lower alkyl are methyl, ethyl, propyl, isopropyl, butyl, isobutyl and tert-butyl.
Lower haloalkyl: the term "lower haloalkyl" refers to C substituted with one or more halogen atoms1-4Straight-chain or branched-chain alkyl.
Optionally substituted: as described herein, the compounds of the present disclosure may contain optionally substituted and/or substituted moieties. In general, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituents may be the same or different at each position. In some embodiments, the optionally substituted group is unsubstituted. The combinations of substituents contemplated by the present disclosure are preferably those that result in the formation of stable or chemically feasible compounds. As used herein, the term "stable" means that the compound is not substantially altered when subjected to conditions that allow its production, detection, and, in certain embodiments, its recovery, purification, and use for one or more of the purposes disclosed herein. Certain substituents are described below.
Suitable monovalent substitutions on substitutable atoms, e.g. suitable carbon atomsIndependently, the radicals are halogen; - (CH)2)0- 4R^;-(CH2)0-4OR^;-O(CH2)0-4Ro;-O-(CH2)0-4C(O)ORo;-(CH2)0-4CH(OR^)2(ii) a Can be substituted by RoSubstituted- (CH)2)0-4Ph; can be substituted by RoSubstituted- (CH)2)0-4O(CH2)0-1Ph; can be substituted by Rosubstituted-CH ═ CHPh; can be substituted by RoSubstituted- (CH)2)0-4O(CH2)0-1-a pyridyl group; -NO2;-CN;-N3;-(CH2)0-4N(R^)2;-(CH2)0-4N(R^)C(O)R^;-N(R^)C(S)R^;-(CH2)0-4N(R^)C(O)NR^2;-N(R^)C(S)NR^2;-(CH2)0-4N(R^)C(O)OR^;-N(R^)N(R^)C(O)R^;-N(R^)N(R^)C(O)NR^2;-N(R^)N(R^)C(O)OR^;-(CH2)0-4C(O)R^;-C(S)R^;-(CH2)0-4C(O)OR^;-(CH2)0-4C(O)SR^;-(CH2)0-4C(O)OSiR^3;-(CH2)0-4OC(O)R^;-OC(O)(CH2)0-4SRo;-SC(S)SRo;-(CH2)0-4SC(O)R^;-(CH2)0-4C(O)NR^2;-C(S)NR^2;-C(S)SRo;-(CH2)0-4OC(O)NR^2;-C(O)N(OR^)R^;-C(O)C(O)R^;-C(O)CH2C(O)R^;-C(NOR^)R^;-(CH2)0-4SSR^;-(CH2)0-4S(O)2R^;-(CH2)0-4S(O)2OR^;-(CH2)0-4OS(O)2R^;-S(O)2NR^2;-(CH2)0-4S(O)R^;-N(R^)S(O)2NR^2;-N(R^)S(O)2R^;-N(OR^)R^;-C(NH)NR^2;-Si(R^)3;-OSi(R^)3;-B(R^)2;-OB(R^)2;-OB(OR^)2;-P(R^)2;-P(OR^)2;-P(R^)(OR^);-OP(R^)2;-OP(OR^)2;-OP(R^)(OR^);-P(O)(R^)2;-P(O)(OR^)2;-OP(O)(R^)2;-OP(O)(OR^)2;-OP(O)(OR^)(SR^);-SP(O)(R^)2;-SP(O)(OR^)2;-N(R^)P(O)(R^)2;-N(R^)P(O)(OR^)2;-P(R^)2[B(R^)3];-P(OR^)2[B(R^)3];-OP(R^)2[B(R^)3];-OP(OR^)2[B(R^)3];-(C1-4Straight or branched alkylene) O-N (R ^)2(ii) a Or- (C)1-4Straight or branched chain alkylene) C (O) O-N (R ^ R)2Wherein each R ^ can be substituted as defined herein and independently is: hydrogen, C1-20Aliphatic radical, C having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, sulfur, silicon and phosphorus1-20Heteroaliphatic group, -CH2-(C6-14Aryl group, -O (CH)2)0-1(C6-14Aryl group), -CH2- (5-to 14-membered heteroaryl ring), a 5-to 20-membered monocyclic, bicyclic or polycyclic saturated, partially unsaturated or aryl ring having 0 to 5 heteroatoms independently selected from nitrogen, oxygen, sulfur, silicon and phosphorus; or in addition to the above definitions, two independently occurring R ^ s together with their intervening atoms form a 5 to 20 membered, monocyclic, bicyclic, or polycyclic saturated, partially unsaturated, or aryl ring having 0 to 5 heteroatoms independently selected from nitrogen, oxygen, sulfur, silicon, and phosphorus, which may be substituted as defined below.
Suitable monovalent substituents on R ^ (or rings formed by combining two independently occurring R ^ with their intervening atoms) are independently halogen, - (CH) 2)0-2R·- (halogeno radical R)·)、-(CH2)0-2OH、-(CH2)0-2OR·、-(CH2)0- 2CH(OR·)2-O (halo R)·)、-CN、-N3、-(CH2)0-2C(O)R·、-(CH2)0-2C(O)OH、-(CH2)0-2C(O)OR·、-(CH2)0-2SR·、-(CH2)0-2SH、-(CH2)0-2NH2、-(CH2)0-2NHR·、-(CH2)0-2NR· 2、-NO2、-SiR· 3、-OSiR· 3、-C(O)SR·、-(C1-4Straight OR branched chain alkylene) C (O) OR·or-SSR·Wherein each R·Unsubstituted or substituted with only one or more halogens when preceded by a "halo" group and is independently selected from: c1-4Aliphatic radical, -CH2Ph,-O(CH2)0-1Ph, or a 5 to 6 membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Suitable divalent substituents on the saturated carbon atom of R ^ include ═ O and ═ S.
For example, suitable divalent substituents on suitable carbon atoms are independently the following: is one of O, S and NNR* 2、=NNHC(O)R*、=NNHC(O)OR*、=NNHS(O)2R*、=NR*、=NOR*、-O(C(R* 2))2-3O-or-S (C (R)* 2))2-3S-, wherein each independently occurs R*Selected from: hydrogen, C which may be substituted as defined below1-6An aliphatic group, or an unsubstituted 5-to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. Suitable divalent substituents bound to adjacent substitutable carbons of the "optionally substituted" group include: -O (CR)* 2)2-3O-, in which each occurrence of R is independent*Selected from: hydrogen, C which may be substituted as defined below1-6An aliphatic group, and an unsubstituted 5-to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
R*Suitable substituents on the aliphatic radical of (a) are independently halogen, -R·- (halogeno radical R)·)、-OH、-OR·-O (halo R)·)、-CN、-C(O)OH、-C(O)OR·、-NH2、-NHR·、-NR· 2Or-NO2Wherein each R·Unsubstituted or substituted with only one or more halogens when preceded by a "halo" group and independently: c1-4Aliphatic radical, -CH2Ph,-O(CH2)0-1Ph, or a 5 to 6 membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, suitable substituents on the substitutable nitrogen are independently
Figure BDA0003526174380000231
Figure BDA0003526174380000232
Or
Figure BDA0003526174380000233
Wherein each one of
Figure BDA0003526174380000234
Independently are: hydrogen, C which may be substituted as defined below1-6An aliphatic group, unsubstituted-OPh, or an unsubstituted 5-to 6-membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or, in addition to the above definitions, two independently
Figure BDA0003526174380000235
Together with their central atoms, form an unsubstituted 3-to 12-membered saturated, partially unsaturated or aryl monocyclic or bicyclic ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen and sulfur.
Figure BDA0003526174380000236
Suitable substituents on the aliphatic radical of (a) are independently halogen, -R·- (halogeno radical R)·)、-OH、-OR·-O (halo R)·)、-CN、-C(O)OH、-C(O)OR·、-NH2、-NHR·、-NR· 2or-NO2Wherein each R·Is not taken outSubstituted or when the foregoing is "halo" is substituted with only one or more halogens and independently is: c 1-4Aliphatic radical, -CH2Ph,-O(CH2)0-1Ph, or a 5 to 6 membered saturated, partially unsaturated, or aryl ring having 0 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
Partial unsaturation: as used herein, the term "partially unsaturated" refers to a cyclic moiety that contains at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to encompass aryl or heteroaryl moieties as defined herein.
The pharmaceutical composition comprises: as used herein, the term "pharmaceutical composition" refers to an active agent formulated with one or more pharmaceutically acceptable carriers. In some embodiments, the active agent is present in a treatment regimen in a unit dose amount suitable for administration that, when administered to a relevant population, exhibits a statistically significant probability of achieving the intended therapeutic effect. In some embodiments, the pharmaceutical composition may be specifically formulated for administration in solid or liquid form, including pharmaceutical compositions suitable for: oral administration, such as drenches (aqueous or non-aqueous solutions or suspensions), tablets (e.g., tablets targeted for buccal, sublingual and systemic absorption), boluses, powders, granules, pastes for application to the tongue; parenteral administration, for example by subcutaneous, intramuscular, intravenous or epidural injection, as for example a sterile solution or suspension or a sustained release formulation; topical application, such as creams, ointments or controlled release patches or sprays applied to the skin, lungs or oral cavity; intravaginally or intrarectally, e.g., as a pessary, cream or foam; under the tongue; eye passing; percutaneous; or nasal, pulmonary, and other mucosal surfaces.
Pharmaceutically acceptable: as used herein, the phrase "pharmaceutically acceptable" refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
A pharmaceutically acceptable carrier: as used herein, the term "pharmaceutically acceptable carrier" means a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, or solvent encapsulating material, involved in carrying or transporting the subject compound from one organ or portion of the body to another organ or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials that can serve as pharmaceutically acceptable carriers include: sugars such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols such as glycerol, sorbitol, mannitol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; no pyrogen water; isotonic saline; ringer's solution (Ringer's solution); ethanol; a pH buffer solution; polyesters, polycarbonates and/or polyanhydrides; as well as other non-toxic compatible materials employed in pharmaceutical formulations.
Pharmaceutically acceptable salts: as used herein, the term "pharmaceutically acceptable salt" refers to salts of such compounds that are suitable for use in a pharmaceutical setting, i.e., salts that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without excessive toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, s.m. berge et al, journal of pharmaceutical Sciences (j. pharmaceutical Sciences), 66: pharmaceutically acceptable salts are described in detail in 1-19 (1977). In some embodiments, pharmaceutically acceptable salts include, but are not limited to, non-toxic acid addition salts of amino groups with inorganic or organic acidsSalts formed with acids, inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid, organic acids such as acetic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid, or salts formed by using other methods used in the art, such as ion exchange methods. In some embodiments, pharmaceutically acceptable salts include, but are not limited to, adipates, alginates, ascorbates, aspartates, benzenesulfonates, benzoates, bisulfates, borates, butyrates, camphorates, camphorsulfonates, citrates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, formates, fumarates, glucoheptonates, glycerophosphates, gluconates, hemisulfates, heptanoates, hexanoates, hydroiodides, 2-hydroxy-ethanesulfonates, lactobionates, lactates, laurates, laurylsulfates, malates, maleates, malonates, methanesulfonates, 2-naphthalenesulfonates, nicotinates, nitrates, oleates, oxalates, palmitates, pamoate, pectinates, persulfates, 3-phenylpropionates, persulfates, salts of alginic acid, salts of citric acid, salts of fumaric acid, salts of gluconic acid, salts of fumaric acid, salts of bifunctional salts of fumaric acid, and salts of maleic acid, salts of bifunctional salts of maleic acid, salts of bifunctional salts of maleic acid, and salts of bifunctional acid, and salts of bifunctional acid, and derivatives of bifunctional salts of bifunctional acid, and of bifunctional salts of bifunctional acid, and use of bifunctional salts of bifunctional acids, and use of bifunctional salts of bifunctional acids, and use of bifunctional salts of bifunctional acids, and of bifunctional salts of bifunctional, Phosphates, picrates, pivalates, propionates, stearates, succinates, sulfates, tartrates, thiocyanates, p-toluenesulfonates, undecanoates, valerates, and the like. In some embodiments, provided compounds include one or more acidic groups, and the pharmaceutically acceptable salt is an alkali metal salt, alkaline earth metal salt, or ammonium salt (e.g., n (r)) 3Wherein each R is independently defined and described in the present disclosure). Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. In some embodiments, the pharmaceutically acceptable salt is a sodium salt. In some embodiments, the pharmaceutically acceptable salt is a potassium salt. In some embodiments, the pharmaceutically acceptable salt is a calcium salt. In some embodiments, pharmaceutically acceptable salts include, where appropriate, non-toxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, alkyl groups having 1 to 6 carbon atoms, sulfonate, and arylsulfonate. In some embodiments, provided compounds include more than one acidA sex group. In some embodiments, a pharmaceutically acceptable salt or, generally, a salt of such a compound includes two or more cations that may be the same or different. In some embodiments, in pharmaceutically acceptable salts (or salts in general), all of the ionizable hydrogens in the acidic groups (e.g., in aqueous solution having a pKa of no more than about 11, 10, 9, 8, 7, 6, 5, 4, 3, or 2; in some embodiments, no more than about 7; in some embodiments, no more than about 6; in some embodiments, no more than about 5; in some embodiments, no more than about 4; in some embodiments, no more than about 3) are replaced with cations.
Protecting groups: as used herein, the term "Protecting group" is well known in the art and includes Protecting Groups described in detail in Organic Synthesis (Protecting Groups in Organic Synthesis), t.w.greene and p.g.m.wuts, 3 rd edition, john wili father publications, 1999, the entire contents of which are incorporated herein by reference. Also included are protecting groups specifically adapted for nucleoside and nucleotide chemistries as described in the Current Nucleic Acid Chemistry protocol (Current Protocols in Nucleic Acid Chemistry) edited by Serge l.beaucage et al 06/2012, the entire contents of chapter 2 of which are incorporated herein by reference. Suitable amino protecting groups include methyl carbamate, ethyl carbamate, 9-fluorenylmethyl carbamate (Fmoc), 9- (2-sulfo) fluorenylmethyl carbamate, 9- (2, 7-dibromo) fluorenylmethyl carbamate, 2, 7-di-tert-butyl- [9- (10, 10-dioxo-10, 10, 10, 10-tetrahydrothioxanthyl) ] methyl carbamate (DBD-Tmoc), 4-methoxybenzoyl methyl carbamate (Phenoc), 2, 2, 2-trichloroethyl carbamate (Troc), 2-trimethylsilylethyl carbamate (Teoc), 2-phenylethyl carbamate (hZ), 1- (1-adamantyl) -1-methylethyl carbamate (Adpoc), 1-carbamic, 1-dimethyl-2-haloethyl carbamate, 1-dimethyl-2, 2-dibromoethyl carbamate (DB-t-BOC), 1-dimethyl-2, 2, 2-trichloroethyl carbamate (TCBOC), 1-methyl-1- (4-biphenylyl) ethyl carbamate (Bpoc), 1- (3, 5-di-tert-butylphenyl) -1-methylethyl carbamate (t-Bumeoc), 2- (2 '-and 4' -pyridyl) ethyl carbamate (Pyoc), 2- (N, N-dicyclohexylcarboxamido) ethyl carbamate, tert-butyl carbamate (BOC), 1-adamantyl carbamate (Adoc), vinyl carbamate (Voc), Allyl carbamate (Alloc), 1-isopropylallyl carbamate (Ipaoc), cinnamyl carbamate (Coc), 4-nitrocinnamyl carbamate (Noc), 8-quinolinyl carbamate, N-hydroxypiperidinyl carbamate, alkyldithio carbamate, benzyl carbamate (Cbz), p-methoxybenzyl carbamate (Moz), p-nitrobenzyl carbamate, p-bromobenzyl carbamate, p-chlorobenzyl carbamate, 2, 4-dichlorobenzyl carbamate, 4-methylsulfinylbenzyl carbamate (Msz), 9-anthracylmethyl carbamate, diphenylmethyl carbamate, 2-methylthioethyl carbamate, 2-methylsulfonylethyl carbamate, 2- (p-toluenesulfonyl) ethyl carbamate, methyl carbamate, ethyl carbamate, N-butyl acetate, N-butyl, N-butyl, N-butyl, N-butyl, [2- (1, 3-dithianyl) ] methyl carbamate (Dmoc), 4-methylthiophene carbamate (Mtpc), 2, 4-dimethylthienyl carbamate (Bmpc), 2-phosphonium ethyl carbamate (Peoc), 2-triphenylphosphonium isopropyl carbamate (Ppoc), 1-dimethyl-2-cyanoethyl carbamate, m-chloro-p-acyloxybenzyl carbamate, p- (dihydroxyoxyboro) benzyl carbamate, 5-benzisoxazolyl methyl carbamate, 2- (trifluoromethyl) -6-chromonylmethyl carbamate (Tcroc), m-nitrophenyl carbamate, 3, 5-dimethoxybenzyl carbamate, o-nitrophenyl carbamate, 3, 4-dimethoxy-6-nitrobenzyl carbamate, 2- (trifluoromethyl) -6-chromonyl methyl carbamate (Tcroc), Phenyl (o-nitrophenyl) methyl carbamate, phenothiazinyl- (10) -carbonyl derivative, N ' -p-toluenesulfonylaminocarbonyl derivative, N ' -phenylaminothiocarbonyl derivative, tert-amyl carbamate, S-benzyl thiocarbamate, p-cyanobenzylcarbamate, cyclobutyl carbamate, cyclohexyl carbamate, cyclopentyl carbamate, cyclopropylmethyl carbamate, p-decyloxybenzyl carbamate, 2-dimethoxycarbonylvinyl carbamate, o- (N, N-dimethylcarboxamido) benzyl carbamate, 1-dimethyl-3- (N, N-dimethylcarboxamido) propyl carbamate, 1-dimethylpropynyl carbamate, di- (2-pyridyl) methyl carbamate, di- (N, N-methyl) methyl carbamate, di- (N, N-methyl) carbamate, di- (2-methyl) carbamate, di- (N, N-methyl) carbamate, di (N-methyl) carbamate, N ' -phenyl-methyl) carbamate, di (N-methyl) carbamate, di (2, N-methyl) carbamate, di (N, di-methyl) carbamate, di-methyl, di-or di-methyl carbamate, 2-furylmethyl carbamate, 2-iodoethyl carbamate, isobornyl carbamate, isobutyl carbamate, isonicotinite carbamate, p- (p' -methoxyphenylazo) benzyl carbamate, 1-methylcyclobutyl carbamate, 1-methylcyclohexyl carbamate, 1-methyl-1-cyclopropylmethyl carbamate, 1-methyl-1- (3, 5-dimethoxyphenyl) ethyl carbamate, 1-methyl-1- (p-phenylazophenyl) ethyl carbamate, 1-methyl-1-phenylethyl carbamate, 1-methyl-1- (4-pyridyl) ethyl carbamate, phenyl carbamate, p- (phenylazo) benzyl carbamate, p-methyl-1- (4-pyridyl) ethyl carbamate, p-methyl-1- (phenylazo) benzyl carbamate, p-methyl-1- (3, 5-dimethoxyphenyl) carbamate, p-methyl-1- (p-phenylazophenyl) ethyl carbamate, n-butyl-methyl-1-cyclopropylmethyl carbamate, n-methyl-1- (4-pyridyl) ethyl carbamate, p-methyl-phenylazo) ethyl carbamate, n-methyl-1-ethyl carbamate, n-methyl-1-methyl-carbamate, n-methyl-1-methyl-carbamate, p-methyl-1-methyl-2-methyl-carbamate, p-methyl-2-methyl-carbamate, and-methyl-ethyl, or-methyl-2-ethyl, or-methyl-ethyl-methyl-ethyl, or-ethyl-methyl-ethyl, or-methyl-ethyl-methyl-ethyl, or-methyl-ethyl carbamate, or-ethyl-methyl-2-methyl-2-ethyl, or-methyl-ethyl, or-methyl-ethyl, or-methyl-or-methyl-or-methyl-ethyl, or-methyl-or-ethyl, or-p-or-p-, 2, 4, 6-tri-tert-butylphenyl carbamate, 4- (trimethylammonium) benzyl carbamate, 2, 4, 6-trimethylbenzyl carbamate, formamide, acetamide, chloroacetamide, trichloroacetamide, trifluoroacetamide, phenylacetamide, 3-phenylpropionamide, picolinamide, 3-pyridylcarboxamide, N-benzoylphenylalanyl derivative, benzamide, p-phenylbenzamide, o-nitrophenylacetamide, o-nitrophenyloxyacetamide, acetoacetamide, (N' -dithiobenzyloxycarbonylamino) acetamide, 3- (p-hydroxyphenyl) propionamide, 3- (o-nitrophenyl) propionamide, 2-methyl-2- (o-nitrophenyloxy) propionamide, 2-methyl-2- (o-phenylazoylphenoxy) propionamide, N-phenylthiocarbonylamino-methyl-3- (p-hydroxyphenyl) propionamide, N-benzoylphenylalanyl derivative, benzamide, p-phenylbenzamide, o-phenylacetamide, o-nitrophenyl-acetamide, o-2-methyl-2- (o-phenylazoyl-phenoxy) propionamide, N-phenylazoyl-propionamide, N-phenyl-3- (p-phenylazoyl) -propionamide, N-phenylazoyl-propionamide, N-phenylazophenoxy) amide, N-propionamide, N-substituted phenyl-acylamide, N-substituted acylamino-substituted acylamino-substituted acylamino, and their derivatives, 4-chlorobutanamide, 3-methyl-3-nitrobutanamide, o-nitrocinnamamide, N-acetylmethionine derivatives, o-nitrobenzamide, o- (benzoyloxymethyl) benzamide, 4, 5-diphenyl-3-oxazolin-2-one, N-phthalimide, N-dithiasuccinimide (Dts), N-2, 3-diphenylmaleimide, N-2, 5-dimethylpyrrole, N-1, 1, 4, 4-tetramethyldiazacyclopentane adduct (STABASE), 5-substituted 1, 3-dimethyl-1, 3, 5-triazacyclohexan-2-one, 5-substituted 1, 3-benzhydryl-1, 3, 5-triazacyclohex-2-one, 1-substituted 3, 5-dinitro-4-pyridone, N-methylamine, N-allylamine, N- [2- (trimethylsilyl) ethoxy ] methylamine (SEM), N-3-acetoxypropylamine, N- (1-isopropyl-4-nitro-2-oxo-3-pyrrolin-3-yl) amine, quaternary ammonium salts, N-benzylamine, N-bis (4-methoxyphenyl) methylamine, N-5-dibenzocycloheptylamine, N-triphenylmethylamine (Tr), N- [ (4-methoxyphenyl) diphenylmethyl ] amine (MMTr), N-9-phenylfluorenylamine (PhF), N-2, 7-dichloro-9-fluorenylmethylidene amine, N-ferrocenylmethylamino (Fcm), N-2-pyridylmethylamino N '-oxide, N-1, 1-dimethylthiomethyl amine, N-benzylidene amine, N-p-methoxybenzylidene amine, N-diphenylmethylidene amine, N- [ (2-pyridyl) mesityl ] methylidene amine, N- (N', N '-dimethylaminomethylene) amine, N' -isopropylidene diamine, N-p-nitrophenylylidene amine, N-salicylidene amine, N-5-chlorosalicylidene amine, N- (5-chloro-2-hydroxyphenyl) phenylmethylidene amine, N-cyclohexylidene amine, N-tolylidene, N- (5, 5-dimethyl-3-oxo-1-cyclohexenyl) amine, N-borane derivatives, N-diphenylboronic acid derivatives, N- [ phenyl (chromium or tungsten pentacarbonyl) carbonyl ] amine, N-copper chelates, N-zinc chelates, N-nitroamines, N-nitrosamines, amine N-oxides, diphenylphosphinamides (Dpp), dimethylthiophosphamides (Mpt), diphenylphosphinamides (Ppt), dialkyl phosphoramidates, dibenzyl phosphoramidates, diphenyl phosphoramidates, benzenesulfenamides, o-nitrobenzenesulfinamides (Nps), 2, 4-dinitrobenzenesulfenamides, pentachlorobenzenesulfinamides, 2-nitro-4-methoxybenzenesulfinamides, triphenylmethylsulfinamides, 3-nitropyridinesulfinamides (Npys), P-toluenesulfonamide (Ts), benzenesulfonamide, 2, 3, 6-trimethyl-4-methoxybenzenesulfonamide (Mtr), 2, 4, 6-trimethoxybenzenesulfonamide (Mtb), 2, 6-dimethyl-4-methoxybenzenesulfonamide (Pme), 2, 3, 5, 6-tetramethyl-4-methoxybenzenesulfonamide (Mte), 4-methoxybenzenesulfonamide (Mbs), 2, 4, 6-trimethylbenzenesulfonamide (Mts), 2, 6-dimethoxy-4-methylbenzenesulfonamide (iMds), 2, 5, 7, 8-pentamethylchroman-6-sulfonamide (Pmc), methanesulfonamide (Ms), β -trimethylsilylethanesulfonamide (SES), 9-anthracenesulfonamide, 4- (4', 8' -Dimethoxynaphthylmethyl) benzenesulfonamide (DNMBS), benzylsulfonamide, trifluoromethylsulfonamide and benzoylmethanesulfonamide.
Suitable protected carboxylic acids further include, but are not limited to, carboxylic acids protected by silane groups, alkyl groups, alkenyl groups, aryl groups, and arylalkyl groups. Examples of suitable silane groups include trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triisopropylsilyl, and the like. Examples of suitable alkyl groups include methyl, benzyl, p-methoxybenzyl, 3, 4-dimethoxybenzyl, trityl, tert-butyl, tetrahydropyran-2-yl. Examples of suitable alkenyl groups include allyl. Examples of suitable aryl groups include optionally substituted phenyl, biphenyl or naphthyl. Examples of suitable arylalkyl groups include optionally substituted benzyl groups (e.g., p-methoxybenzyl (MPM), 3, 4-dimethoxybenzyl, O-nitrobenzyl, p-halophenyl-methyl, 2, 6-dichlorobenzyl, p-cyanobenzyl) as well as 2-picolyl and 4-picolyl.
Suitable hydroxy protecting groups include methyl, methoxymethyl (MOM), methylthiomethyl (MTM), tert-butylthiomethyl, (phenyldimethylsilyl) methoxymethyl (SMOM), Benzyloxymethyl (BOM), p-methoxyphenylmethoxymethyl (PMBM), (4-methoxyphenoxy) methyl (p-AOM), Guaiacolmethyl (GUM), tert-butoxymethyl, 4-Pentenyloxymethyl (POM), silanyloxymethyl, 2-methoxyethoxymethyl (MEM), 2, 2, 2-trichloroethoxymethyl, bis (2-chloroethoxy) methyl, 2- (trimethylsilyl) ethoxymethyl (SEMOR), Tetrahydropyranyl (THP), 3-bromotetrahydropyranyl, tetrahydrothiopyranyl, 1-methoxycyclohexyl, 4-Methoxytetrahydropyranyl (MTHP), 4-methoxytetrahydrothiopyranyl, 4-methoxytetrahydrothiopyranyl S, S-dioxide, 1- [ (2-chloro-4-methyl) phenyl ] -4-methoxypiperidin-4-yl (CTMP), 1, 4-dioxan-2-yl, tetrahydrofuranyl, tetrahydrothiofuranyl, 2, 3, 3a, 4, 5, 6, 7, 7 a-octahydro-7, 8, 8-trimethyl-4, 7-methanobenzofuran-2-yl, 1-ethoxyethyl, 1- (2-chloroethoxy) ethyl, 1-methyl-1-methoxyethyl, 1-methyl-1-benzyloxyethyl, 1-methyl-1-benzyloxy-2-fluoroethyl, 2, 2, 2-trichloroethyl, 2-trimethylsilylethyl, 2- (phenylseleno) ethyl, tert-butyl, allyl, p-chlorophenyl, p-methoxyphenyl, 2, 4-dinitrophenyl, benzyl, p-methoxybenzyl, 3, 4-dimethoxybenzyl, o-nitrobenzyl, p-halogenobenzyl, 2, 6-dichlorobenzyl, p-cyanobenzyl, p-phenylbenzyl, 2-picolyl, 4-picolyl, 3-methyl-2-picolyl N-oxido, diphenylmethyl, p' -dinitrobenzhydryl, 5-dibenzosuberyl, triphenylmethyl, alpha-naphthyldiphenylmethyl, p-methoxyphenyldiphenylmethyl, di (p-methoxyphenyl) phenylmethyl, di (p-methoxyphenyl) phenyl methyl, di (p-N-oxido) methyl, p-halogenobenzyl, p-halobenzyl, p-nitrobenzyl, p-cyanobenzyl, p-phenoxybenzyl, p-trifluoromethylphenyl, p-phenoxybenzyl, p-nitrophenyl, p-trifluoromethylphenyl, p-nitrophenyl, p-iodobenzyl, p-iodophenyl, p-iodophenyl, p-iodophenyl, iodo, Tris (p-methoxyphenyl) methyl, 4- (4 ' -bromobenzoyloxyphenyl) diphenylmethyl, 4 ' -tris (4, 5-dichlorophthalimidophenyl) methyl, 4 ' -tris (levulinoyloxyphenyl) methyl, 4 ' -tris (benzoyloxyphenyl) methyl, 3- (imidazol-1-yl) bis (4 ', 4 ' -dimethoxyphenyl) methyl, 1-bis (4-methoxyphenyl) -1 ' -pyrenylmethyl, 9-anthryl, 9- (9-phenyl) xanthenyl, 9- (9-phenyl-10-oxo) anthryl, 1, 3-benzodithiolan-2-yl, benzisothiazolyl S, s-dioxyionic group, trimethylsilyl group (TMS), triethylsilyl group (TES), triisopropylsilyl group (TIPS), dimethylisopropylsilyl group (IPDMS), diethylisopropylsilyl group (DEIPS), dimethyl tert-hexylsilyl group, tert-butyldimethylsilyl group (TBDMS), tert-butyldiphenylsilyl group (TBDPS), tritylsilyl group, tri-p-xylylsilyl group, triphenylsilyl group, diphenylmethylsilyl group (DPMS), tert-butylmethoxyphenylsilyl group (TBMPS), formate ester, benzoylformate ester, acetate ester, chloroacetate ester, dichloroacetate ester, trichloroacetate ester, trifluoroacetate ester, methoxyacetate ester, triphenylmethoxyacetate ester, phenoxyacetate ester, p-chlorophenoxyacetate ester, 3-phenylpropionate ester, 4-oxopentanoate ester (levulinate ester), 4, 4- (ethylenedithio) valerate (levulinyl dithioacetal), pivalate, adamantane ester, crotonate, 4-methoxycrotonate, benzoate, p-phenylbenzoate, 2, 4, 6-trimethylbenzoate (mesitylate), alkyl carbonate methyl ester, 9-fluorenylmethyl carbonate (Fmoc), alkyl carbonate ethyl ester, alkyl 2, 2, 2-trichloroethyl carbonate (Troc), 2- (trimethylsilyl) ethyl carbonate (TMSEC), 2- (phenylsulfonyl) ethyl carbonate (Psec), 2- (triphenylphosphonio) ethyl carbonate (Peoc), alkyl isobutyl carbonate, alkyl vinyl carbonate, allyl carbonate, alkyl p-nitrophenyl carbonate, alkyl benzyl carbonate, alkyl p-methoxybenzyl carbonate, allyl carbonate, alkyl p-nitrophenyl carbonate, methyl, Alkyl carbonate 3, 4-dimethoxybenzyl ester, alkyl carbonate o-nitrophenyl ester, alkyl carbonate p-nitrophenyl ester, alkyl thiocarbonate S-benzyl ester, 4-ethoxy-1-naphthyl carbonate, methyl dithiocarbonate, 2-iodobenzoate, 4-azidobutyrate, 4-nitro-4-methylpentanoate, o- (dibromomethyl) benzoate, 2-formylbenzenesulfonate, 2- (methylthiomethoxy) ethyl, 4- (methylthiomethoxy) butyrate, 2- (methylthiomethoxymethyl) benzoate, 2, 6-dichloro-4-methylphenoxyacetate, 2, 6-dichloro-4- (1, 1, 3, 3-tetramethylbutyl) phenoxyacetate, p-toluenesulfonate, iodobenzoate, 2, 6-iodomethyl-4-methylphenoxyacetate, 2, 6-chloro-4-phenoxyacetate, 2, 6-chloro-4- (1, 3, 3-tetramethylbutyl) phenoxyacetate, p-toluenesulfonate, p-phenoxyacetate, p-toluenesulfonate, p-iodobenzoate, and/p-phenoxyacetate, p-iodobenzoate, p-phenoxyacetate, p-iodobenzoate, p-phenoxyacetate, p-iodobenzoate, and/p-iodobenzoate, p-phenoxyacetate, p-iodobenzoate, p-phenoxyacetate, p-iodobenzoate, and/p-iodobenzoate, 2, 4-bis (1, 1-dimethylpropyl) phenoxyacetate, chlorodiphenylacetate, isobutyrate, monobutyrate, (E) -2-methyl-2-butenoate, o- (methoxycarbonyl) benzoate, α -naphthoate, nitrate, alkyl N, N, N ', N' -tetramethylphosphorodiamidate, alkyl N-phenylcarbamate, borate, dimethylphosphinylsulfinyl, alkyl 2, 4-dinitrophenylsulfinate, sulfate, methanesulfonate (methanesulfonate/mesylate), benzylsulfonate, and tosylate (Ts). For protecting 1, 2-diols or 1, 3-diols, the protective group comprises methylene acetal, ethylene acetal, 1-tert-butylethylene ketal, 1-phenylethylene ketal, (4-methoxyphenyl) ethylene acetal, 2, 2, 2-trichloroethylene acetal, acetonide, cyclopentylene ketal, cyclohexylene ketal, cycloheptylene ketal, benzylidene acetal, p-methoxybenzylidene acetal, 2, 4-dimethoxybenzylidene ketal, 3, 4-dimethoxybenzylidene acetal, 2-nitrobenzylidene acetal, methoxymethylene acetal, ethoxymethylene acetal, dimethoxymethylene orthoester, 1-methoxyethylene orthoester, 1-ethoxyethylene orthoester, 1, 2-dimethoxyethylene orthoester, 1, 3-dimethoxyethylene orthoester, α -methoxybenzylidene orthoester, 1- (N, N-dimethylamino) ethylene derivative, α - (N, N' -dimethylamino) benzylidene derivative, 2-oxacyclopentylidene orthoester, di-t-butylsilylene (DTBS), 1, 3- (1, 1, 3, 3-tetraisopropyldisiloxane) derivative (TIPDS), tetra-t-butoxydisiloxane-1, 3-diylidene derivative (TBDS), cyclic carbonate, cyclic borate, ethyl borate, and phenyl borate.
In some embodiments, the hydroxyl protecting group is acetyl, t-butyl, t-butoxymethyl, methoxymethyl, tetrahydropyranyl, 1-ethoxyethyl, 1- (2-chloroethoxy) ethyl, 2-trimethylsilylethyl, p-chlorophenyl, 2, 4-dinitrophenyl, benzyl, benzoyl, p-phenylbenzoyl, 2, 6-dichlorobenzyl, diphenylmethyl, p-nitrobenzyl, triphenylmethyl (trityl), 4' -dimethoxytrityl, trimethylsilyl, triethylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, triphenylsilyl, triisopropylsilyl, benzoylformate, chloroacetyl, trichloroacetyl, trifluoroacetyl, pivaloyl, 9-fluorenylmethyl carbonate, p-tert-butyldiphenylsilyl, and mixtures thereof, Mesylate, tosylate, triflate, trityl, monomethoxytrityl (MMTr), 4 '-dimethoxytrityl (DMTr) and 4, 4' -trimethoxytrityl (TMTr), 2-cyanoethyl (CE or Cne), 2- (trimethylsilyl) ethyl (TSE), 2- (2-nitrophenyl) ethyl, 2- (4-cyanophenyl) ethyl, 2- (4-nitrophenyl) ethyl (NPE), 2- (4-nitrophenylsulfonyl) ethyl, 3, 5-dichlorophenyl, 2, 4-dimethylphenyl, 2-nitrophenyl, 4-nitrophenyl, 2, 4, 6-trimethylphenyl, 2- (2-nitrophenyl) ethyl, butylthiocarbonyl, 4, 4' -tris (benzoyloxy) trityl, diphenylcarbamoyl, acetylpropyl, 2- (dibromomethyl) benzoyl (Dbmb), 2- (isopropylthiomethoxymethyl) benzoyl (Ptmt), 9-phenylxanthen-9-yl (pixyl) or 9- (p-methoxyphenyl) xanth-9-yl (MOX). In some embodiments, each of the hydroxyl protecting groups is independently selected from acetyl, benzyl, tert-butyldimethylsilyl, tert-butyldiphenylsilyl, and 4, 4' -dimethoxytrityl. In some embodiments, the hydroxyl protecting group is selected from the group consisting of: trityl, monomethoxytrityl and 4, 4' -dimethoxytrityl. In some embodiments, a phosphorus bond protecting group is a group that is attached to a phosphorus bond (e.g., an internucleotide bond) during the entire oligonucleotide synthesis. In some embodiments, a protecting group is attached to the sulfur atom of the phosphorothioate group. In some embodiments, the protecting group is attached to an oxygen atom of an internucleotide phosphorothioate linkage. In some embodiments, the protecting group is attached to an oxygen atom of an internucleotide phosphate linkage. In some embodiments, the protecting group is 2-cyanoethyl (CE or Cne), 2-trimethylsilylethyl, 2-nitroethyl, 2-sulfonylethyl, methyl, benzyl, o-nitrobenzyl, 2- (p-nitrophenyl) ethyl (NPE or Npe), 2-phenylethyl, 3- (N-tert-butylcarboxamido) -1-propyl, 4-oxopentyl, 4-methylthio-1-butyl, 2-cyano-1, 1-dimethylethyl, 4-N-methylaminobutyl, 3- (2-pyridyl) -1-propyl, 2- [ N-methyl-N- (2-pyridyl) ] aminoethyl, 2- (N-formyl, n-methyl) aminoethyl or 4- [ N-methyl-N- (2, 2, 2-trifluoroacetyl) amino ] butyl.
Subject: as used herein, the term "subject" refers to any organism to which a compound or composition is administered, e.g., for experimental, diagnostic, prophylactic and/or therapeutic purposes, according to the present disclosure. Typical subjects include animals (e.g., mammals such as mice, rats, rabbits, non-human primates, and humans; insects; worms, etc.) and plants. In some embodiments, the subject is a human. In some embodiments, the subject may be suffering from and/or susceptible to a disease, disorder, and/or condition.
Essentially: as used herein, the term "substantially" refers to a qualitative condition that exhibits a total or near total range or degree of a feature or characteristic of interest. One of ordinary skill in the art will appreciate that biological and chemical phenomena rarely, if ever, go to completion and/or proceed to completion or achieve or avoid absolute results. Thus, the term "substantially" is used herein to capture the lack of potential completeness inherent in many biological and/or chemical phenomena.
Susceptible to: an individual "susceptible to" a disease, disorder, and/or condition is one who is at a higher risk of developing the disease, disorder, and/or condition than a member of the general public. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition is predisposed to the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not have been diagnosed with the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition may not exhibit symptoms of the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition will develop the disease, disorder, and/or condition. In some embodiments, an individual who is susceptible to a disease, disorder, and/or condition does not develop the disease, disorder, and/or condition.
Therapeutic agents: as used herein, the term "therapeutic agent" generally refers to any agent that, when administered to a subject, elicits a desired effect (e.g., a desired biological, clinical, or pharmaceutical effect). In some embodiments, an agent is considered a therapeutic agent if it exhibits a statistically significant effect in the appropriate population. In some embodiments, a suitable population is a population of subjects suffering from and/or susceptible to a disease, disorder, or condition. In some embodiments, the appropriate population is a population of model organisms. In some embodiments, the appropriate population may be defined by one or more criteria, such as age group, gender, genetic background, pre-existing clinical condition, prior exposure to therapy. In some embodiments, a therapeutic agent is a substance that, when administered to a subject in an effective amount, alleviates, ameliorates, inhibits, prevents, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms or features of a disease, disorder, and/or condition in the subject. In some embodiments, a "therapeutic agent" is a medicament that has been or requires approval by a governmental agency for sale to humans for administration. In some embodiments, a "therapeutic agent" is a medicament that requires a medical prescription to be administered to a human. In some embodiments, the therapeutic agent is a compound described herein.
A therapeutically effective amount of: as used herein, the term "therapeutically effective amount" means an amount of a substance (e.g., a therapeutic agent, composition, and/or formulation) that elicits a desired biological response when administered as part of a treatment regimen. In some embodiments, a therapeutically effective amount of a substance is an amount sufficient to treat, diagnose, prevent, and/or delay the onset of a disease, disorder, and/or condition when administered to a subject suffering from or susceptible to such a disease, disorder, and/or condition. As will be appreciated by those skilled in the art, the effective amount of a substance may vary depending on factors such as the desired biological indicator, the substance to be delivered, the target cell or tissue, and the like. For example, an effective amount of a compound in a formulation for treating a disease, disorder, and/or condition is an amount that alleviates, ameliorates, reduces, inhibits, prevents, delays onset of, reduces severity of, and/or reduces incidence of one or more symptoms or features of the disease, disorder, and/or condition. In some embodiments, the therapeutically effective amount is administered in a single dose; in some embodiments, multiple unit doses are required to deliver a therapeutically effective amount.
Treatment: as used herein, the terms "treat", "treating" or "treating" refer to any method for partially or completely alleviating, ameliorating, alleviating, inhibiting, preventing, delaying the onset of, reducing the severity of, and/or reducing the incidence of one or more symptoms or features of a disease, disorder, and/or condition. The treatment can be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition. In some embodiments, treatment may be administered to a subject who exhibits only early signs of a disease, disorder, and/or condition, for example, for the purpose of reducing the risk of developing a pathology associated with the disease, disorder, and/or condition.
Unit dose: the expression "unit dose" as used herein refers to an amount administered in a single dose and/or physically discrete units of a pharmaceutical composition. In many embodiments, the unit dose contains a predetermined amount of active agent. In some embodiments, a unit dose contains an entire single dose of a pharmaceutical agent. In some embodiments, more than one unit dose is administered to achieve a total single dose. In some embodiments, it is necessary or expected that multiple unit doses will need to be administered in order to achieve the intended effect. A unit dose can be, for example, a volume of liquid (e.g., an acceptable carrier) containing a predetermined amount of one or more therapeutic agents, a predetermined amount of one or more therapeutic agents in solid form, a sustained release formulation or drug delivery device containing a predetermined amount of one or more therapeutic agents, or the like. It will be appreciated that the unit dose can be presented in a formulation that includes any of a variety of components in addition to the therapeutic agent. For example, acceptable carriers (e.g., pharmaceutically acceptable carriers), diluents, stabilizers, buffers, preservatives, and the like can be included, as described below. It will be appreciated by those skilled in the art that in many embodiments, the total appropriate daily dose of a particular therapeutic agent may comprise a fraction or multiple unit doses and may be determined, for example, by an attending physician within the scope of sound medical judgment. In some embodiments, a particular effective dosage level for any particular subject or organism may depend upon a variety of factors, including the disorder being treated and the severity of the disorder; the activity of the particular active compound used; the particular composition used; the age, weight, general health, sex, and diet of the subject; the time of administration and the rate of excretion of the particular active compound used; the duration of treatment; drugs and/or other therapies used in combination or concomitantly with the particular compound employed; and similar factors well known in the medical arts.
Unsaturated reaction: as used herein, the term "unsaturated" means a moiety having one or more units of unsaturation.
Unless otherwise indicated, the structures depicted herein are also intended to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms of the structures described; for example, R and S conformations, Z and E double bond isomers, and Z and E conformational isomers for each asymmetric center. Thus, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the disclosed compounds are within the scope of the disclosure. Unless otherwise indicated, all tautomeric forms of the compounds are within the scope of the disclosure. In addition, unless otherwise indicated, structures depicted herein are also intended to encompass the presence of only one in the structureOr a plurality of isotopically enriched atoms. For example, involving replacement of hydrogen by deuterium or tritium or by13C or14Carbon-enriched carbon-substituted compounds having the structure of the present disclosure are within the scope of the present disclosure. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents according to the present disclosure. As will be appreciated by those skilled in the art, the provided compounds, agents, etc. may be provided in the form of solvates thereof.
3. Description of the exemplary embodiments:
in some embodiments, the present disclosure provides a medicament comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
optionally a linker moiety.
In some embodiments, the antibody binding moiety is uABT. In some embodiments, the target binding moiety may bind to CD 38. In some embodiments, the agent is a compound of formula I, I-a, I-b, II, or III, or a salt thereof. In some embodiments, the present disclosure provides a compound of formula I, I-a, I-b, II, or III, or a pharmaceutically acceptable salt thereof. Various embodiments of the provided technology are described herein as examples.
Antibody binding moieties
The present disclosure provides, inter alia, agents comprising antibody binding moieties, such as ARM. In some embodiments, the antibody binding moiety is a universal antibody binding moiety that can bind to antibodies having different Fab regions and different specificities. In some embodiments, the antibody binding portion of the present disclosure is a universal antibody binding portion that binds to an Fc region. In some embodiments, binding of the universal antibody binding moiety to the Fc region may occur simultaneously with binding of an Fc receptor (e.g., CD16a) to the same Fc region (e.g., may be at different positions/amino acid residues of the same Fc region). In some embodiments, upon binding a universal antibody binding moiety, e.g., in provided agents, compounds, methods, etc., the Fc region may still interact with the Fc receptor and perform one or more or all of its immune activities, including recruiting immune cells (e.g., effector cells, e.g., NK cells), and/or triggering, generating, promoting, and/or enhancing immune system activities, e.g., antibody-dependent cell-mediated cytotoxicity (ADCC) and/or ADCP, against a target cell, tissue, subject, and/or entity.
Various universal antibody binding moieties may be utilized in accordance with the present disclosure. Certain antibody binding moieties and techniques for identifying and/or assessing generic antibody binding moieties and/or their use in ARM are described in WO/2019/023501 and are incorporated herein by reference. Those skilled in the art will appreciate that additional techniques in the art may be suitable for identifying and/or assessing universal antibody binding moieties suitable for an ARM according to the present disclosure. In some embodiments, the universal antibody binding moiety comprises one or more amino acid residues, each of which is independently natural or non-natural. In some embodiments, the universal antibody binding moiety has
Figure BDA0003526174380000341
Structure (la) or a salt form thereof. In some embodiments, the universal antibody binding moiety has
Figure BDA0003526174380000342
Structure (la) or a salt form thereof. In some embodiments, the universal antibody binding moiety is or includes a peptide moiety, e.g., having Rc(Xaa) a moiety of the z-structure or a salt form thereof, wherein RcEach of z and Xaa is independently as described herein. In some embodiments, one or more Xaa are independently an unnatural amino acid residue. In some embodiments, the side chains of two or more amino acid residues may be linked together to form a bridge. For example, in some embodiments, the side chains of two cysteine residues may form a disulfide bridge comprising-S- (as in many proteins, it may be formed by two-SH groups). In some embodiments, the universal antibody binding moiety is or includes a cyclic peptide moiety, e.g., having
Figure BDA0003526174380000343
Of the structure or its salt formsAnd (4) partial. In some embodiments, the universal antibody binding moiety is Rc- (Xaa) z-or
Figure BDA0003526174380000344
Or a salt form thereof, and is or includes a peptide unit. In some embodiments, - (Xaa) z-is or includes a peptide unit. In some embodiments, the peptide unit comprises an amino acid residue (e.g., at physiological pH of about 7.4, a "positively charged amino acid residue" XaaP) For example, residues of amino acids of the formulae A-I having positively charged side chains. In some embodiments, the peptide unit comprises R. In some embodiments, at least one Xaa is R. In some embodiments, the peptide unit is or includes an APAR. In some embodiments, the peptide unit is or comprises RAPA. In some embodiments, the peptide unit comprises an amino acid residue, e.g., a residue of an amino acid of formula a-I having a side chain comprising an aromatic group ("aromatic amino acid residue" Xaa)A). In some embodiments, the peptide unit includes a positively charged amino acid residue and an aromatic amino acid residue. In some embodiments, the peptide unit comprises W. In some embodiments, the peptide unit includes a positively charged amino acid residue and an aromatic amino acid residue. In some embodiments, the peptide unit is or comprises XaaAXaaXaaPXaaP. In some embodiments, the peptide unit is or comprises Xaa PXaaPXaaXaaA. In some embodiments, the peptide unit is or comprises XaaPXaaAXaaP. In some embodiments, the peptide unit is or comprises two or more XaaPXaaAXaaP. In some embodiments, the peptide unit is or comprises XaaPXaaAXaaPXaaXaaPXaaAXaaP. In some embodiments, the peptide unit is or comprises XaaPXaaPXaaAXaaAXaaP. In some embodiments, the peptide unit is or comprises XaaPXaaPXaaPXaaA. In some embodiments, the peptide unit is or comprises two or more XaaAXaaAXaaP. In some embodiments, the peptide residue comprises one or more prolinesAn acid residue. In some embodiments, the peptide unit is or comprises HWRGWA. In some embodiments, the peptide unit is or comprises WGRR. In some embodiments, the peptide unit is or comprises RRGW. In some embodiments, the peptide unit is or comprises NKFRGKYK. In some embodiments, the peptide unit is or comprises NRFRGKYK. In some embodiments, the peptide unit is or comprises NARKFYK. In some embodiments, the peptide unit is or comprises NARKFYKG. In some embodiments, the peptide unit is or comprises HWRGWV. In some embodiments, the peptide unit is or comprises KHFRNKD. In some embodiments, a peptide unit includes positively charged amino acid residues, aromatic amino acid residues, and amino acid residues, such as the residues of amino acids of formulae a-I having a negatively charged side chain (e.g., at physiological pH of about 7.4, "negatively charged amino acid residue" Xaa) N). In some embodiments, the peptide residue is RHRFNKD. In some embodiments, the peptide unit is TY. In some embodiments, the peptide unit is TYK. In some embodiments, the peptide unit is RTY. In some embodiments, the peptide unit is RTYK. In some embodiments, the peptide unit is or comprises a sequence selected from PAM. In some embodiments, the peptide unit is WHL. In some embodiments, the peptide unit is ELVW. In some embodiments, the peptide unit is or includes a sequence selected from AWHLGELVW. In some embodiments, the peptide unit is or includes a sequence selected from DCAWHLGELVWCT in which two cysteine residues may form a disulfide bond, as seen in native proteins. In some embodiments, the peptide unit is or includes a sequence selected from Fc-III. In some embodiments, the peptide unit is or includes a sequence selected from DpLpAWHLGELVW. In some embodiments, the peptide unit is or includes a sequence selected from FcBP-1. In some embodiments, the peptide unit is or comprises a sequence selected from dplpdcahlglcvwct. In some embodiments, the peptide unit is or includes a sequence selected from FcBP-2. In some embodiments, the peptide unit is or includes a sequence selected from CDCAWHLGELVWCTC, wherein the first and last cysteines and the two cysteines in the middle of the sequence may each independently form a disulfide bond as in the native protein. In some embodiments, the peptide unit is or includes a sequence selected from Fc-III-4 c. In some embodiments, the peptide unit is or comprises an FcRM selected from And (4) sequencing. In some embodiments, the peptide unit is or comprises a cyclic peptide unit. In some embodiments, the cyclic peptide unit includes an amide group formed by an amino group of the side chain and the C-terminal-COOH.
In some embodiments, - (Xaa) z-is or includes [ X1]p1[X2]p2-X3X4X5X6X7X8X9X10X11X12-[X13]p13-[X14]p14[X15]p15[X16]p16Wherein X is1、X2、X3、X4、X5、X6、X7、X8、X9、X10、X11、X12And X13Each of which is independently an amino acid residue, e.g., an amino acid of formula a-I, and each of p1, p2, p13, p14, p15, and p16 is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In some embodiments, X1、X2、X3、X4、X5、X6、X7、X8、X9、X10、X11、X12And X13Each of which is independently an amino acid residue of an amino acid of formula a-I. In some embodiments, X1、X2、X3、X4、X4、X6、X7、X8、X9、X10、X11、X12And X13Each of which is independently a natural amino acid residue. In some embodiments, X1、X2、X3、X4、X5、X6、X7、X8、X9、X10、X11、X12And X13Is an unnatural amino acid residue as described in the disclosure.
In some embodiments, the peptide unit includes a functional group of an amino acid residue that is reactive with a functional group of another amino acid residue. In some embodiments, the peptide unit comprises a peptide havingAn amino acid residue of a side chain comprising a functional group that can react with another functional group of a side chain of another amino acid residue to form a bond (see, e.g., the moieties described in table a-1, table 1, etc.). In some embodiments, one functional group of one amino acid residue is linked to a functional group of another amino acid residue to form a bond (or bridge). The bond is to the backbone atom of the peptide unit and does not include the backbone atom. In some embodiments, a peptide unit comprises a bond formed by two side chains of non-adjacent amino acid residues. In some embodiments, the bond is bonded to two backbone atoms of two non-adjacent amino acid residues. In some embodiments, both backbone atoms bonded to the bond are carbon atoms. In some embodiments, the key has L bStructure of, wherein LbIs L as described in this disclosureaWherein L isaNot a covalent bond. In some embodiments, Laincluding-Cy-. In some embodiments, Laincluding-Cy-wherein-Cy-is optionally substituted heteroaryl. In some embodiments, -Cy-is
Figure BDA0003526174380000361
In some embodiments, LaIs composed of
Figure BDA0003526174380000362
In some embodiments, such LaCan be derived from the side chain of an amino acid residue3Formation of a group and a side chain of another amino acid residue. In some embodiments, the bond is formed via the attachment of two thiol groups, e.g., two cysteine residues. In some embodiments, Laincluding-S-S-. In some embodiments, Lais-CH2-S-S-CH2-. In some embodiments, the bond is via an amino group (e.g., -NH in the side chain of a lysine residue)2) And a carboxylic acid group (e.g., -COOH in the side chain of an aspartic acid or glutamic acid residue). In some embodiments, Lacomprising-C (O) -N (R') -. In some embodiments, Laincluding-C (O) -NH-. In some embodiments, Lais-CH2CONH-(CH2)3-。In some embodiments, Laincluding-C (O) -N (R ') -, where R' is R, and forms a ring with the R group on the peptide backbone (e.g., as in A-34). In some embodiments, LaIs- (CH)2)2-N(R′)-CO--(CH2)2-. In some embodiments, -Cy-is optionally substituted phenylene. In some embodiments, -Cy-is optionally substituted 1, 2-phenylene. In some embodiments, L aIs composed of
Figure BDA0003526174380000363
In some embodiments, LaIs composed of
Figure BDA0003526174380000364
In some embodiments, LaIs optionally substituted divalent C2-20A divalent aliphatic group. In some embodiments, LaIs optionally substituted- (CH)2)9-CH=CH-(CH2)9-. In some embodiments, LaIs- (CH)2)3-CH=CH-(CH2)3-。
In some embodiments, the two amino acid residues bonded to the bond are separated by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more than 15 amino acid residues (not including the two amino acid residues bonded to the bond). In some embodiments, the number is 1. In some embodiments, the number is 2. In some embodiments, the number is 3. In some embodiments, the number is 4. In some embodiments, the number is 5. In some embodiments, the number is 6. In some embodiments, the number is 7. In some embodiments, the number is 8. In some embodiments, the number is 9. In some embodiments, the number is 10. In some embodiments, the number is 11. In some embodiments, the number is 12. In some embodiments, the number is 13. In some embodiments, the number is 14. In some embodiments, the number is 15.
In some embodiments, each of p1, p2, p13, p14, p15, and p16 is 0. In some embodiments, - (Xaa) z-is or includes-X 3X4X5X6X7X8X9X10X11X12-, wherein:
X3、X4、X5、X6、X7、X8、X9、X10、X11and X12Each of which is independently an amino acid residue;
X6is XaaAOr XaaP
X9Is XaaN(ii) a And is
X12Is XaaAOr XaaP
In some embodiments, X3、X4、X5、X6、X7、X8、X9、X10、X11And X12Each of which is independently an amino acid residue of an amino acid of formula a-I as described in the present disclosure. In some embodiments, X5Is XaaAOr XaaP. In some embodiments, X5Is XaaA. In some embodiments, X5Is XaaP. In some embodiments, X5Is an amino acid residue whose side chain includes an optionally substituted saturated, partially saturated or aromatic ring. In some embodiments, X5Is composed of
Figure BDA0003526174380000371
In some embodiments, X5Is composed of
Figure BDA0003526174380000372
In some embodiments, X6Is XaaA. In some embodiments, X6Is XaaP. In some embodiments, X6Is His. In some embodiments, X12Is XaaA. In some embodiments, X12Is XaaP. In some embodiments, X9Is Asp. In some embodiments, X9Is Glu. In some embodiments, X12Is composed of
Figure BDA0003526174380000373
In some embodiments, X12Is composed of
Figure BDA0003526174380000374
In some embodiments, X7、X10And X11Each of which is independently an amino acid residue having a hydrophobic side chain (a "hydrophobic amino acid residue" Xaa)H). In some embodiments, X7Is XaaH. In some embodiments, X 7Is composed of
Figure BDA0003526174380000375
In some embodiments, X7Is Val. In some embodiments, X10Is XaaH. In some embodiments, X10Is Met. In some embodiments, X10Is composed of
Figure BDA0003526174380000376
In some embodiments, X11Is XaaH. In some embodiments, X11Is composed of
Figure BDA0003526174380000377
In some embodiments, X8Is Gly. In some embodiments, X4Is Pro. In some embodiments, X3Is Lys. In some embodiments, X12of-COOH with Lys (X)3) The side chain amino group of (A) forms an amide bond, and Lys (X)3) Is linked to a linker moiety and then to a target binding moiety.
In some embodiments, - (Xaa) z-is or includes-X3X4X5X6X7X8X9X10X11X12-, wherein:
X3、X4、X5、X6、X7、X8、X9、X10、X11and X12Each of which is independently an amino acid residueA group;
at least two amino acid residues via one or more bonds LbConnecting;
Lbis selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution wherein L isb(ii) is bonded to, and does not include, a backbone atom of one amino acid residue and a backbone atom of another amino acid residue;
X6is XaaAOr XaaP
X9Is XaaN(ii) a And is
X12Is XaaAOr XaaP
In some embodiments, X3、X4、X5、X6、X7、X8、X9、X10、X11And X12Each of which is independently an amino acid residue of an amino acid of formula a-I as described in the present disclosure. In some embodiments, two non-adjacent amino acid residues are through LbAnd (4) connecting. In some embodiments, X5And X10Through LbAnd (4) connecting. In some embodiments, there is one key Lb. In some embodiments, X6Is XaaA. In some embodiments, X6Is XaaP. In some embodiments, X6Is His. In some embodiments, X9Is Asp. In some embodiments, X9Is Glu. In some embodiments, X12Is XaaA. In some embodiments, X12Is composed of
Figure BDA0003526174380000381
In some embodiments of the present invention, the,X12is composed of
Figure BDA0003526174380000382
In some embodiments, X12Is composed of
Figure BDA0003526174380000383
In some embodiments, X4、X7And X11Each of which is independently XaaH. In some embodiments, X4Is XaaH. In some embodiments, X4Is Ala. In some embodiments, X7Is XaaH. In some embodiments, X7Is composed of
Figure BDA0003526174380000384
In some embodiments, X11Is XaaH. In some embodiments, X11Is composed of
Figure BDA0003526174380000385
In some embodiments, X8Is Gly. In some embodiments, X3Is Lys. In some embodiments, X12of-COOH with Lys (X)3) The side chain amino group of (A) forms an amide bond, and Lys (X)3) Is linked to a linker moiety and then to a target binding moiety. In some embodiments, L bIs composed of
Figure BDA0003526174380000391
In some embodiments, LbIs composed of
Figure BDA0003526174380000392
In some embodiments, LbTwo alpha-carbon atoms connecting two different amino acid residues. In some embodiments, X5And X10Are both Cys, and the two-SH groups of their side chains form-S-S- (L)bis-CH2-S-S-CH2-)。
In some embodiments, - (Xaa) z-is or includes-X2X3X4X5X6X7X8X9X10X11X12-, wherein:
X2、X3、X4、X5、X6、X7、X8、X9、X10、X11and X12Each of which is independently an amino acid residue;
at least two amino acid residues via one or more bonds LbConnecting;
Lbis selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution wherein L isb(ii) is bonded to, and does not include, a backbone atom of one amino acid residue and a backbone atom of another amino acid residue;
X4is XaaA
X5Is XaaAOr XaaP
X8Is XaaN(ii) a And is
X11Is XaaA
In some embodiments, X2、X3、X4、X5、X6、X7、X8、X9、X10、X11And X12Each of which is independently an amino acid residue of an amino acid of formula a-I as described in the present disclosure. In some embodiments, two non-adjacent amino acid residues are through LbAnd (4) connecting. In some embodiments, there is one key Lb. In some embodiments, X 2And X12Through LbAnd (4) connecting. In some embodiments, Lbis-CH2-S-S-CH2-. In some embodimentsIn, Lbis-CH2-CH2-S-CH2-. In some embodiments, LbIs composed of
Figure BDA0003526174380000393
In some embodiments, LbIs composed of
Figure BDA0003526174380000394
In some embodiments, Lbis-CH2CH2CO-N(R′)-CH2CH2-. In some embodiments, R 'and-N (R') -CH2CH2The R groups on the backbone atoms to which they are bonded together form a ring, for example as in A-34. In some embodiments, the formed ring is 3-, 4-, 5-, 6-, 7-, or 8-membered. In some embodiments, the ring formed is monocyclic. In some embodiments, the formed ring is saturated. In some embodiments, LbIs composed of
Figure BDA0003526174380000401
In some embodiments, LbTwo alpha-carbon atoms connecting two different amino acid residues. In some embodiments, X4Is XaaA. In some embodiments, X4Is Tyr. In some embodiments, X5Is XaaA. In some embodiments, X5Is XaaP. In some embodiments, X5Is His. In some embodiments, X8Is Asp. In some embodiments, X8Is Glu. X11Is Tyr. In some embodiments, X2And X12Are both Cys, and the two-SH groups of their side chains form-S-S- (L)bis-CH2-S-S-CH2-). In some embodiments, X3、X6、X9And X10Each of which is independently Xaa H. In some embodiments, X3Is XaaH. In some embodiments, X3Is Ala. In some embodiments, X6Is XaaH. In some embodiments, X6Is Leu. In some embodiments, X9Is XaaH. In some embodiments, X9Is Leu. In some embodiments, X9Is composed of
Figure BDA0003526174380000402
In some embodiments, X10Is XaaH. In some embodiments, X10Is Val. In some embodiments, X10Is composed of
Figure BDA0003526174380000403
In some embodiments, X7Is Gly. In some embodiments, p1 is 1. In some embodiments, X1Is Asp. In some embodiments, p13 is 1. In some embodiments, p14, p15, and p16 are 0. In some embodiments, X13Is an amino acid residue comprising a polar uncharged side chain (e.g., at physiological pH, "polar uncharged amino acid residue" XaaL). In some embodiments, X13Is Thr. In some embodiments, X13Is Val. In some embodiments, p13 is 0. In some embodiments, Rcis-NHCH2CH(OH)CH3. In some embodiments, RcIs (R) -NHCH2CH(OH)CH3. In some embodiments, RcIs (S) -NHCH2CH(OH)CH3
In some embodiments, - (Xaa) z-is or includes-X2X3X4X5X6X7X8X9X10X11X12-, wherein:
X2、X3、X4、X5、X6、X7、X8、X9、X10、X11and X12Each of which is independently an amino acid residue;
at least two amino acid residues via one or more bonds L bConnecting;
Lbis selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20Heteroaliphatic groupOptionally substituted divalent radical of (a) wherein one or more methylene units of said radical are optionally and independently substituted by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution wherein L isb(ii) is bonded to, and does not include, a backbone atom of one amino acid residue and a backbone atom of another amino acid residue;
X5is XaaAOr XaaP
X8Is XaaN(ii) a And is
X11Is XaaA
In some embodiments, X2、X3、X4、X5、X6、X7、X8、X9、X10、X11And X12Each of which is independently an amino acid residue of an amino acid of formula a-I as described in the present disclosure. In some embodiments, two non-adjacent amino acid residues are through LbAnd (4) connecting. In some embodiments, there is one key Lb. In some embodiments, there are two or more bonds Lb. In some embodiments, there are two keys Lb. In some embodiments, X2And X12Through LbAnd (4) connecting. In some embodiments, X4And X9Through LbAnd (4) connecting. In some embodiments, X4And X10Through LbAnd (4) connecting. In some embodiments, Lbis-CH2-S-S-CH2-. In some embodiments, LbIs composed of
Figure BDA0003526174380000411
In some embodiments, LbIs composed of
Figure BDA0003526174380000412
In some embodiments, X2And X12Both Cys and two of their side chains with-SH groups forming-S- (L)bis-CH2-S-S-CH2-). In some embodiments, X4And X10Are both Cys, and the two-SH groups of their side chains form-S-S- (L)bis-CH2-S-S-CH2-). In some embodiments, X4And X9Through LbIs connected, wherein LbIs composed of
Figure BDA0003526174380000413
In some embodiments, X4And X9Through LbIs connected, wherein LbIs composed of
Figure BDA0003526174380000414
In some embodiments, X5Is XaaA. In some embodiments, X5Is XaaP. In some embodiments, X5Is His. In some embodiments, X8Is Asp. In some embodiments, X8Is Glu. In some embodiments, X11Is Tyr. In some embodiments, X11Is composed of
Figure BDA0003526174380000415
In some embodiments, X2And X12Through LbIs connected, wherein Lbis-CH2-S-CH2CH2-. In some embodiments, LbTwo alpha-carbon atoms connecting two different amino acid residues. In some embodiments, X3、X6And X9Each of which is independently XaaH. In some embodiments, X3Is XaaH. In some embodiments, X3Is Ala. In some embodiments, X6Is XaaH. In some embodiments, X6Is Leu. In some embodiments, X6Is composed of
Figure BDA0003526174380000416
In some embodiments, X9Is XaaH. In some embodiments, X9Is Leu. In some casesIn the examples, X9Is composed of
Figure BDA0003526174380000417
In some embodiments, X10Is XaaH. In some embodiments, X 10Is Val. In some embodiments, X7Is Gly. In some embodiments, p1 is 1. In some embodiments, X1Is XaaN. In some embodiments, X1Is Asp. In some embodiments, X1Is Glu. In some embodiments, p13 is 1. In some embodiments, p14, p15, and p16 are 0. In some embodiments, X13Is XaaL. In some embodiments, X13Is Thr. In some embodiments, X13Is Val.
In some embodiments, - (Xaa) z-is or includes-X2X3X4X5X6X7X8X9X10X11X12X13X14X15X16-, wherein:
X2、X3、X4、X5、X6、X7、X8、X9、X10、X11、X12、X13、X14、X15and X16Each of which is independently an amino acid residue;
at least two amino acid residues via a bond LbConnecting;
Lbis selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution wherein L isbBound to the main chain atom of one amino acid residue and to the main chain atom of another amino acid residueChain atoms, and not including backbone atoms;
X3is XaaN
X6Is XaaA
X7Is XaaAOr XaaP
X9Is XaaN(ii) a And is
X13Is XaaA
In some embodiments, X2、X3、X4、X5、X6、X7、X8、X9、X10、X11And X12Each of which is independently an amino acid residue of an amino acid of formula a-I as described in the present disclosure. In some embodiments, two non-adjacent amino acid residues are through L bAnd (4) connecting. In some embodiments, there is one key Lb. In some embodiments, there are two or more bonds Lb. In some embodiments, there are two keys Lb. In some embodiments, X2Through LbIs connected to X16. In some embodiments, X4Through LbIs connected to X14. In some embodiments, X2And X16Are both Cys, and the two-SH groups of their side chains form-S-S- (L)bis-CH2-S-S-CH2-). In some embodiments, X4And X14Are both Cys, and the two-SH groups of their side chains form-S-S- (L)bis-CH2-S-S-CH2-). In some embodiments, LbTwo alpha-carbon atoms connecting two different amino acid residues. In some embodiments, X3Is Asp. In some embodiments, X3Is Glu. In some embodiments, X5Is XaaH. In some embodiments, X5Is Ala. In some embodiments, X6Is XaaA. In some embodiments, X6Is Tyr. In some embodiments, X7Is XaaA. In some embodiments, X7Is XaaP. In some embodiments, X7Is His. At one endIn some embodiments, X8Is XaaH. In some embodiments, X8Is Ala. In some embodiments, X9Is Gly. In some embodiments, X10Is Asp. In some embodiments, X10Is Glu. In some embodiments, X 11Is XaaH. In some embodiments, X11Is Leu. In some embodiments, X12Is XaaH. In some embodiments, X12Is Val. In some embodiments, X13Is XaaA. In some embodiments, X13Is Tyr. In some embodiments, X15Is XaaL. In some embodiments, X15Is Thr. In some embodiments, X15Is Val. In some embodiments, p1 is 1. In some embodiments, X1Is XaaN. In some embodiments, X1Is Asp. In some embodiments, X1Is Glu.
As will be appreciated by those skilled in the art, an amino acid residue may be replaced by another amino acid residue having similar properties, e.g., XaaH(e.g., Val, Leu, etc.) may be substituted with another XaaH(e.g., Leu, Ile, Ala, etc.) substitution, a XaaACan be substituted by another XaaAReplacement, one XaaPCan be substituted by another XaaPReplacement, one XaaNCan be substituted by another XaaNReplacement, one XaaLCan be substituted by another XaaLPermutation, and the like.
In some embodiments, the antibody binding moiety (e.g., a universal antibody binding moiety) is or includes an optionally substituted moiety of table a-1 in some embodiments, the antibody binding moiety (e.g., a universal antibody binding moiety) is selected from table a-1.
TABLE A-1 exemplary antibody binding moieties.
Figure BDA0003526174380000431
Figure BDA0003526174380000441
Figure BDA0003526174380000451
Figure BDA0003526174380000461
Figure BDA0003526174380000471
Figure BDA0003526174380000481
Figure BDA0003526174380000491
Figure BDA0003526174380000501
Figure BDA0003526174380000511
Figure BDA0003526174380000521
In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-1. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-2. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-3. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-4. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-5. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-6. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-7. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-8. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-9. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-10. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-11. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-12. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-13. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-14. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-15. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-16. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-17. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-18. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-19. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-20. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-21. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-22. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-23. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-24. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-25. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-26. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-27. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-28. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-29. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-30. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-31. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-32. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-33. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-34. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-35. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-36. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-37. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-38. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-39. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-40. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-41. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-42. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-43. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-44. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-45. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-46. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-47. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-48. In some embodiments, the universal antibody binding moiety is or includes optionally substituted a-49.
In some embodiments, the universal antibody binding moiety is A-1. In some embodiments, the universal antibody binding moiety is a-2. In some embodiments, the universal antibody binding moiety is a-3. In some embodiments, the universal antibody binding moiety is a-4. In some embodiments, the universal antibody binding moiety is a-5. In some embodiments, the universal antibody binding moiety is a-6. In some embodiments, the universal antibody binding moiety is a-7. In some embodiments, the universal antibody binding moiety is a-8. In some embodiments, the universal antibody binding moiety is a-9. In some embodiments, the universal antibody binding moiety is A-10. In some embodiments, the universal antibody binding moiety is a-11. In some embodiments, the universal antibody binding moiety is a-12. In some embodiments, the universal antibody binding moiety is a-13. In some embodiments, the universal antibody binding moiety is A-14. In some embodiments, the universal antibody binding moiety is a-15. In some embodiments, the universal antibody binding moiety is a-16. In some embodiments, the universal antibody binding moiety is a-17. In some embodiments, the universal antibody binding moiety is A-18. In some embodiments, the universal antibody binding moiety is a-19. In some embodiments, the universal antibody binding moiety is A-20. In some embodiments, the universal antibody binding moiety is a-21. In some embodiments, the universal antibody binding moiety is a-22. In some embodiments, the universal antibody binding moiety is a-23. In some embodiments, the universal antibody binding moiety is A-24. In some embodiments, the universal antibody binding moiety is a-25. In some embodiments, the universal antibody binding moiety is a-26. In some embodiments, the universal antibody binding moiety is a-27. In some embodiments, the universal antibody binding moiety is a-28. In some embodiments, the universal antibody binding moiety is a-29. In some embodiments, the universal antibody binding moiety is A-30. In some embodiments, the universal antibody binding moiety is a-31. In some embodiments, the universal antibody binding moiety is a-32. In some embodiments, the universal antibody binding moiety is a-33. In some embodiments, the universal antibody binding moiety is a-34. In some embodiments, the universal antibody binding moiety is a-35. In some embodiments, the universal antibody binding moiety is A-36. In some embodiments, the universal antibody binding moiety is a-37. In some embodiments, the universal antibody binding moiety is a-38. In some embodiments, the universal antibody binding moiety is a-39. In some embodiments, the universal antibody binding moiety is A-40. In some embodiments, the universal antibody binding moiety is A-41. In some embodiments, the universal antibody binding moiety is A-42. In some embodiments, the universal antibody binding moiety is a-43. In some embodiments, the universal antibody binding moiety is a-44. In some embodiments, the universal antibody binding moiety is a-45. In some embodiments, the universal antibody binding moiety is A-46. In some embodiments, the universal antibody binding moiety is a-47. In some embodiments, the universal antibody binding moiety is a-48. In some embodiments, the universal antibody binding moiety is a-49.
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000551
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000552
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000553
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000561
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000562
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000563
In some embodiments, the antibody binding moiety is or comprises
Figure BDA0003526174380000571
In some embodiments, the universal antibody binding moiety comprises a peptide unit and is linked to the linker moiety via the C-terminus of the peptide unit. In some embodiments, it is linked to the linker moiety via the N-terminus of the peptide unit. In some embodiments, it is linked to the linker via a side chain group of the peptide unit. In some embodiments, the universal antibody binding moiety comprises a peptide unit and is linked to the target binding moiety via the C-terminus of the peptide unit, optionally via a linker moiety. In some embodiments, the universal antibody binding moiety comprises a peptide unit and is linked to the target binding moiety via the N-terminus of the peptide unit, optionally via a linker moiety. In some embodiments, the universal antibody binding moiety comprises a peptide unit and is linked to the target binding moiety via a side chain of the peptide unit, optionally via a linker moiety.
In some embodiments, an antibody binding moiety (e.g., a universal antibody binding moiety) is or includes a small molecule entity with a molecular weight of, for example, less than 10000, 9000, 8000, 7000, 6000, 5000, 4000, 3000, 2000, 1500, 1000, and the like. Suitable such antibody binding moieties comprise small molecule Fc binding agent moieties such as those described in US 9,745,339, US 201/30131321 and the like. In some embodiments, the antibody binding moiety has a structure whose corresponding compound is a compound described in US 9,745,339 or US 2013/0131321, the compounds of each of which are independently incorporated herein by reference. In some embodiments, ABT has the structure of H-ABT is a compound described in US 9,745,339 or US 2013/0131321, the compounds of each of which are independently incorporated herein by reference. In some embodiments, such compounds can bind to antibodies. In some embodiments, such compounds can bind to the Fc region of an antibody.
In some embodiments, the antibody binding moiety, e.g., ABT, is or includes optionally substituted
Figure BDA0003526174380000572
In some embodiments, ABT is or comprises
Figure BDA0003526174380000581
In some embodiments, ABT is or includes optionally substituted
Figure BDA0003526174380000582
In some embodiments, ABT is or comprises
Figure BDA0003526174380000583
In some embodiments, ABT is or includes optionally substituted
Figure BDA0003526174380000584
In some embodiments, ABT is or comprises
Figure BDA0003526174380000585
In some embodiments, ABT is or includes optionally substituted
Figure BDA0003526174380000586
In some embodiments, ABT is or comprises
Figure BDA0003526174380000587
In some embodiments, the antibody binding moiety is a triazine moiety, such as the triazine moiety described in US 2009/0286693. In some embodiments, the antibody binding portion has a structure whose corresponding compound is that described in US 2009/0286693, which compounds are independently incorporated herein by reference. In some embodiments, ABT has the structure of H-ABT is a compound described in US 2009/0286693, which compounds are independently incorporated herein by reference. In some embodiments, such compounds can bind to antibodies. In some embodiments, such compounds can bind to the Fc region of an antibody.
In some embodiments, the antibody binding moiety is a triazine moiety, such as Teng et al, a strategy to generate biomimetic ligands for affinity chromatography (a strategy for the generation of biological ligands for affinity chromatography), Combinatorial synthesis and biological evaluation of IgG binding ligands (Combinatorial synthesis and biological evaluation of an IgG binding ligand), journal of molecular recognition (j.mol.recognition.) 1999; 12: 67-75 ("Teng"). In some embodiments, the antibody-binding portion has a structure whose corresponding compound is that described in Teng, which compounds are independently incorporated herein by reference. In some embodiments, ABT has the structure of H-ABT being a compound described in Teng, which compounds are independently incorporated herein by reference. In some embodiments, such compounds can bind to antibodies. In some embodiments, such compounds can bind to the Fc region of an antibody.
In some embodiments, the antibody binding moiety is a Triazine moiety, such as Uttamchandri et al, a microarray combining a Triazine library for labeling in Small Molecule ligand Discovery of Human IgG (microarray of labeled Combinatorial ligand conjugates in the Discovery of Small-Molecule Ligands of Human IgG), journal of Combinatorial chemistry (J Comb Chem.) 2004 from month 11 to month 12; 6(6): 862-8 ("Uttamchandri"). In some embodiments, the antibody-binding portion has a structure whose corresponding compound is that described in ottamchandani, which compounds are independently incorporated herein by reference. In some embodiments, the ABT has the structure where H-ABT is a compound described in ottamchandani, which compounds are independently incorporated herein by reference. In some embodiments, such compounds can bind to antibodies. In some embodiments, such compounds can bind to the Fc region of an antibody.
In some embodiments, the antibody binding moiety binds to one or more binding sites of protein a. In some embodiments, the antibody binding moiety binds to one or more binding sites of protein G. In some embodiments, the antibody binding moiety binds to one or more binding sites of protein L. In some embodiments, the antibody binding moiety binds to one or more binding sites of protein Z. In some embodiments, the antibody binding moiety binds to one or more binding sites of the protein LG. In some embodiments, the antibody binding moiety binds to one or more binding sites of the protein LA. In some embodiments, the antibody binding moiety binds to one or more binding sites of the protein AG. In some embodiments, the antibody binding moiety is described in Choe, w., Durgannavar, t.a., and Chung, S.J. (2016.) Fc binding ligands of immunoglobulin G: an overview of high affinity proteins and peptides (Fc-binding ligands of immunoglobulin G: An overview of high affinity proteins and peptides) Materials (Materials), 9(12) https:// doi.org/10.3390/ma 9120994.
In some embodiments, the antibody binding moiety can bind to a nucleotide binding site. In some embodiments, the antibody binding moiety is a small molecule moiety that can bind to a nucleotide binding site. In some embodiments, the small molecule is a tryptamine. In some embodiments, the ABT has a structure where H-ABT is tryptamine. Some suitable techniques are described in Mustafaoglu et al, using Affinity Membrane Chromatography methods using Small Molecule targeted Nucleotide Binding sites for antibody Purification (Purification via Affinity Membrane Chromatography Method) with analysts (analysts) 2016, 11/28 days; 141(24): 6571 and 6582.
A number of techniques are available for identifying and/or assessing and/or characterizing antibody binding moieties, including generic antibody binding moieties, and/or their use in ARM, for example the techniques described in WO/2019/023501, the techniques of which are incorporated herein by reference. In some embodiments, the antibody-binding moiety is a moiety (e.g., a small molecule moiety, a peptide moiety, a nucleic acid moiety, etc.) that can selectively bind to IgG and can provide and/or stimulate ADCC and/or ADCP when used in ARM. In some embodiments, the antibody-binding portion can be identified using peptide display techniques (e.g., phase display, non-cell display, etc.). In some embodiments, an antibody-binding moiety is a moiety (e.g., a small molecule moiety, a peptide moiety, a nucleic acid moiety, etc.) that can bind to IgG and optionally can compete with known antibody-binding agents (e.g., protein a, protein G, protein L, etc.).
As will be appreciated by those of skill in the art, antibodies having various properties and activities (e.g., antibodies recognizing different antigens, having optional modifications, etc.) can be recruited through the antibody binding portions described in the present disclosure. In some embodiments, such antibodies comprise an antibody administered to a subject, e.g., for therapeutic purposes. In some embodiments, the antibody recruited by the antibody binding portion comprises an antibody directed against a different antigen. In some embodiments, the antibody recruited by the antibody binding portion comprises an antibody for which the antigen is not present on the surface or cell membrane of a target cell (e.g., a target cell such as a cancer cell). In some embodiments, the antibody recruited by the antibody binding moiety comprises an antibody that does not target an antigen present on the surface or cell membrane of a target (e.g., a target cell, such as a cancer cell). In some embodiments, antigens on the surface of a target cell may interfere with the structure, conformation, and/or one or more properties and/or activities of a recruited antibody that binds such antigen. In some embodiments, as will be appreciated by those of skill in the art, the provided techniques include recruiting a universal antibody binding portion of antibodies with diverse specificities, and no more than 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the recruited antibodies are directed to the same antigen, protein, lipid, carbohydrate, etc. Furthermore, it is an advantage of the present disclosure that the provided technology comprising a universal antibody binding moiety can utilize a diverse pool of antibodies, such as the pool of antibodies present in serum. In some embodiments, a universal antibody binding moiety of the present disclosure (e.g., a universal antibody binding moiety in ARM) is contacted with a plurality of antibodies, wherein no more than 1%, 2%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99% of the plurality of antibodies are directed against the same antigen, protein, lipid, carbohydrate, etc.
CD38
CD38 (differentiation group 38, also known as cyclic ADP ribohydrolases) is expressed by different types of cells and performs multiple functions. It has been reported that in many immune cells, for example CD4+、CD8+CD38 was found on the surface of B lymphocytes and natural killer cells as a glycoprotein. Other functions of CD38, such as cell adhesion, signaling, and calcium signaling, have also been reported. In various embodimentsIn (3), CD38 is human CD 38.
In some embodiments, CD38 is reported to be a non-lineage-restricted type II transmembrane glycoprotein that synthesizes and hydrolyzes cyclic adenosine 5' -diphosphate-ribose (intracellular calcium ion mobilization messenger). It is reported that the release of soluble proteins and the ability of membrane-bound proteins to be internalized may be indicative of both extracellular and intracellular functions of the protein. According to certain reports, CD38 has, in each case, an N-terminal cytoplasmic tail, a single transmembrane domain, and a C-terminal extracellular region with four N-glycosylation sites. Analysis of the crystal structure reportedly indicates that the functional molecule is a dimer, in which the central portion contains the catalytic site. CD38 has been reported to be useful as a prognostic marker for patients with chronic lymphocytic leukemia. Alternative splicing has been reported and can result in a variety of transcriptional variants.
CD38 has been reported to be expressed on immune system cells (e.g., T cells or B cells) of healthy individuals. In some embodiments, in certain conditions, disorders or diseases, an increase in CD38 expression and/or activity levels is observed in cells that do not normally have or have lower levels of CD 38.
CD38 is associated with various conditions, disorders or diseases, such as HIV infection and various cancers, such as leukemia, myeloma, solid tumors, Chronic Lymphocytic Leukemia (CLL), Multiple Myeloma (MM), Acute Promyelocytic Leukemia (APL), non-Hodgkin's lymphoma, B-and T-cell acute lymphocytic leukemia, acute myelogenous leukemia, Hodgkin's lymphoma, chronic myelogenous leukemia, and the like.
Daratumab (an antibody targeting CD 38) has been approved for the treatment of multiple myeloma.
Target binding moieties
Various types and chemical classes of target-binding moieties can be utilized in accordance with the present disclosure. In addition, the compounds of the present disclosure include a target binding moiety that can bind to CD 38. In some embodiments, the target binding moiety binds to a characteristic agent, such as CD 38. In some embodiments, the target binding moiety is or includes a peptide moiety. In some embodiments, the target binding moiety is or includes a nucleic acid agent, such as an aptamer. In some embodiments, the target binding moiety is or comprises a lipid moiety. Certain types of target-binding moieties are described below; those skilled in the art will appreciate that other types of target-binding moieties, including many known in the art, may also be utilized in accordance with the present disclosure. As will be appreciated by those skilled in the art, various techniques are readily available and may be used to assess and confirm CD38 binding. Certain useful techniques are described in the examples.
a. Small molecules
In some embodiments, the target binding moiety is a small molecule moiety. In some embodiments, the small molecule moiety has a molecular weight of no more than 8000, 7000, 6000, 5000, 4000, 3000, 2000, 1500, 1000, 900, 800, 700, or 600. In some embodiments, the small molecule moiety has a molecular weight of no more than 8000. In some embodiments, the small molecule moiety has a molecular weight of no more than 7000. In some embodiments, the small molecule moiety has a molecular weight of no more than 6000. In some embodiments, the small molecule moiety has a molecular weight of no more than 5000. In some embodiments, the small molecule moiety has a molecular weight of no more than 4000. In some embodiments, the small molecule moiety has a molecular weight of no more than 3000. In some embodiments, the small molecule moiety has a molecular weight of no more than 2000. In some embodiments, the small molecule moiety has a molecular weight of no more than 1500. In some embodiments, the small molecule moiety has a molecular weight of no more than 1000. In some embodiments, the small molecule moiety has a molecular weight of no more than 900. Further, the present disclosure encompasses the following recognition: the small molecule target-binding moiety may be capable of binding to a marker, such as CD38, external, on the surface, and/or internal to the target (e.g., cancer cells).
In some embodiments, the small molecule target binding moiety is or includes a moiety that selectively binds to a protein or fragment thereof (e.g., CD 38).
b. Peptide agents
In some embodiments, the target binding moiety is or includes a peptide agent. In some embodiments, the target binding moiety is a peptide moiety. In some embodiments, the peptide moiety can be linear or cyclic. In some embodiments, the target binding moiety is or includes a cyclic peptide moiety. Various peptide target binding moieties are known in the art and can be utilized in accordance with the present disclosure.
In some embodiments, the target binding moiety is or comprises a peptide aptamer agent.
c. Aptamer agents
In some embodiments, the target binding moiety is or comprises a nucleic acid agent. In some embodiments, the target binding moiety is or comprises an oligonucleotide moiety. In some embodiments, the target binding moiety is or comprises an aptamer agent. Various aptamer agents are known in the art or can be readily developed using common techniques, and can be used in the techniques provided in accordance with the present disclosure.
In some embodiments, a target binding moiety, for example, one that can bind to CD38, is or includes a peptide moiety. In some embodiments, the peptide moiety is or includes (Xaa) y or a salt form thereof as described herein. In some embodiments, a target binding moiety, e.g., one that can bind to CD38, is or includes a peptide moiety, e.g., a moiety having the structure:
Figure BDA0003526174380000621
Or a salt thereof, wherein:
each Xaa is independently a residue of an amino acid or amino acid analog;
y is 5 to 20;
LTa linker moiety which is two separate residues of an amino acid or amino acid analogue and is independently a covalent bond, or is selected from C1-C6Aliphatic radical or C having 1 to 5 hetero atoms1-C6A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each RcIndependently is-La-R′;
t is 0 to 50;
each LaIndependently a covalent bond, or is selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from C3-20Cycloaliphatic Ring, C6-20An aryl ring, a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms, and a 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c 1-30Aliphatic radical, C having 1 to 10 heteroatoms1-30Heteroaliphatic radical, C6-30Aryl radical, C6-30Arylaliphatic radical, C having 1 to 10 heteroatoms6-30Aryl heteroaliphatic, 5-to 30-membered heteroaryl having 1 to 10 heteroatoms and 3-to 30-membered heterocyclyl having 1 to 10 heteroatoms, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, together with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the atom; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the intervening atoms.
In some embodiments of the present invention, the,
Figure BDA0003526174380000631
is composed of
Figure BDA0003526174380000632
In some embodiments, each Xaa is independently an amino acid residue. In some embodiments, Xaa is an amino acid analog residue. In some embodiments, one or more Xaa are independently a natural amino acid residue. In some embodiments, one or more Xaa are independently an unnatural amino acid residue. In some embodiments, the side chains of two or more amino acid residues may be linked together to form a bridge. In some embodiments, R cAnd amino acid residue side chains may be linked together to form a bridge. In some embodiments, each bridge independently has La、LbOr LTThe structure of (1). In some embodiments, each bridge independently has LaAnd (5) structure. In some embodiments, each bridge independently has LbAnd (5) structure. In some embodiments, each bridge independently has LTAnd (5) structure. For example, in some embodiments, the side chains of two cysteine residues may form a disulfide bridge comprising-S- (as in many proteins, it may be formed by two-SH groups). In some embodiments, Rcis-La-R, Xaa are residues of amino acids having the structure of formula A-I, wherein Ra2And Ra3One of them is R, and RcR and R ofa2And Ra3Together form a covalent bond.
In some embodiments, - (Xaa) y-is or includes-XaaT1-XaaT2-(Xaa)y′-XaaT3-XaaT4-XaaT5-,
Wherein:
y' is 0 to 8;
XaaT1for C whose side chain is substituted1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT2including an optionally substituted aromatic group for its side chain or being optionally substituted C3-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT3is C whose side chain is optionally substituted2-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT4Including an optionally substituted aromatic group for its side chain or being optionally substituted C3-C8A residue of an amino acid or amino acid analog of an aliphatic group; and is
XaaT5For C whose side chain is substituted1-C8Aliphatic amino acids or amino acid analogues.
In some embodiments, y' is 0. In some embodiments, y' is 1. In some embodiments, y' is 2. In some embodiments, y' is 3. In some embodiments, y' is 4. In some embodiments, y' is 5. In some embodiments, y' is 6. In some embodiments, y' is 7. In some embodiments, y' is 8.
In some embodiments, XaaT1Including substituted C1-C8An aliphatic group. In some embodiments, XaaT1Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, XaaT1Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT1Has a side chain of C2-C8An alkyl group. In some embodiments, XaaT1Is or includes an optionally substituted linear C2-C8An alkyl group. In some embodiments, XaaT1Has a side chain of linear C2-C8An alkyl group. In some embodiments, the side chain is n-pentyl. In some embodiments, Xaa T1Is (S) -NH-CH (n-C)5H11) -C (O) -. In some embodiments, XaaT1Is or includes an aromatic groupAnd (4) clustering. In some embodiments, the side chain is-CH2-R, wherein-CH2-is optionally substituted, and R is optionally substituted aryl or heteroaryl. In some embodiments, the side chain is that of Y, W, S, K or K (MePEG4 c). In some embodiments, the side chain is that of Y, W or S. In some embodiments, XaaT1Is the residue of Y. In some embodiments, XaaT1Is the residue of W. In some embodiments, XaaT1Is the residue of S. In some embodiments, XaaT1Is the residue of K. In some embodiments, XaaT1Is the residue of K (MePEG4 c).
In some embodiments, XaaT2Is or includes an aromatic group. In some embodiments, XaaT2Side chain of (A) is-CH2-R, wherein-CH2-is optionally substituted, and R is as described herein. In some embodiments, R is optionally substituted aryl or heteroaryl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is 4-hydroxyphenyl. In some embodiments, R is 4-phenylphenyl. In some embodiments, the side chain is that of Y or W. In some embodiments, XaaT2Is the residue of Y. In some embodiments, Xaa T2Is the residue of W. In some embodiments, XaaT2Is the residue of Bph. In some embodiments, XaaT2Including substituted C1-C8An aliphatic group. In some embodiments, XaaT2Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, XaaT2Is or includes optionally substituted C3-C8An aliphatic group. In some embodiments, XaaT2Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT2Has a side chain of C2-C8An alkyl group. In some embodiments, XaaT2Is or includes an optionally substituted linear C2-C8An alkyl group. In some embodiments, XaaT2Has a side chain of linear C2-C8An alkyl group. In some embodiments, XaaT2Has a side chain ofC of a branched chain3-C8An alkyl group. In some embodiments, the side chain is n-pentyl. In some embodiments, XaaT2Is (S) -NH-CH (n-C)5H11) -C (O) -. In some embodiments, the side chain is (CH)3)2CHCH2-. In some embodiments, XaaT2Is a residue of L. In some embodiments, XaaT2Is the residue of A.
In some embodiments, XaaT3Including substituted C1-C8An aliphatic group. In some embodiments, XaaT3Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, Xaa T3Is or includes optionally substituted C3-C8An aliphatic group. In some embodiments, XaaT3Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT3Has a side chain of C2-C8An alkyl group. In some embodiments, XaaT3Is or includes an optionally substituted linear C2-C8An alkyl group. In some embodiments, XaaT3Has a side chain of linear C2-C8An alkyl group. In some embodiments, XaaT3The side chain of (A) is branched C3-C8An alkyl group. In some embodiments, the side chain is n-pentyl. In some embodiments, XaaT3Is the residue of Ahp. In some embodiments, the side chain is that of L, V or T. In some embodiments, XaaT3Is a residue of L. In some embodiments, XaaT3Is the residue of V. In some embodiments, XaaT3Is the residue of T. In some embodiments, XaaT3Including inclusion of two or more sp3A side chain of a carbon atom. In some embodiments, XaaT3Comprising a side chain comprising two or more groups, each of said groups independently being-CH2-or-CH3. In some embodiments, XaaT3Including, for example, -OH, -SO2-and the like. In some embodiments, XaaT3Is the residue of Hse (homoserine). In some cases In the examples, XaaT3Is the residue of MetO2 (methionine sulfone).
In some embodiments, XaaT4Is or includes an aromatic group. In some embodiments, XaaT4Side chain of (A) is-CH2-R, wherein-CH2-is optionally substituted, and R is as described herein. In some embodiments, R is optionally substituted aryl or heteroaryl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is 4-hydroxyphenyl. In some embodiments, R is 4-phenylphenyl. In some embodiments, the side chain is that of Y or W. In some embodiments, XaaT4Is the residue of Y. In some embodiments, XaaT4Is the residue of W. In some embodiments, XaaT4Is the residue of Bph. In some embodiments, XaaT4Including substituted C1-C8An aliphatic group. In some embodiments, XaaT4Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, XaaT4Is or includes optionally substituted C3-C8An aliphatic group. In some embodiments, XaaT4Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT4Has a side chain of C2-C8An alkyl group. In some embodiments, XaaT4Is or includes an optionally substituted linear C 2-C8An alkyl group. In some embodiments, XaaT4Has a side chain of linear C2-C8An alkyl group. In some embodiments, XaaT4The side chain of (A) is branched C3-C8An alkyl group. In some embodiments, the side chain is n-pentyl. In some embodiments, the side chain is isopropyl. In some embodiments, XaaT4Is the residue of V. In some embodiments, XaaT4Is the residue of Ahp.
In some embodiments, XaaT5Including substituted C1-C20An aliphatic group. In some embodiments, XaaT5Including substituted C1-C15An aliphatic group. At one endIn some embodiments, XaaT5Including substituted C1-C10An aliphatic group. In some embodiments, XaaT5Including substituted C1-C8An aliphatic group. In some embodiments, XaaT5Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, XaaT5Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT5Has a side chain of C2-C8An alkyl group. In some embodiments, XaaT5Is or includes an optionally substituted linear C2-C8An alkyl group. In some embodiments, XaaT5Has a side chain of linear C2-C8An alkyl group. In some embodiments, the side chain is n-pentyl. In some embodiments, XaaT5Is (S) -NH-CH (n-C)5H11) -C (O) -. In some embodiments, Xaa T5Is or includes an aromatic group. In some embodiments, the side chain is-CH2-R, wherein-CH2-is optionally substituted, and R is optionally substituted aryl or heteroaryl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is 4-phenylphenyl. In some embodiments, the side chain is that of Y, W or S. In some embodiments, XaaT5Is the residue of Y. In some embodiments, XaaT5Is the residue of W. In some embodiments, XaaT5Is the residue of Bph. In some embodiments, XaaT5Is the residue of S. In some embodiments, XaaT5Is the residue of Ado. In some embodiments, XaaT5Is a residue of Ano. In some embodiments, XaaT5Is a residue of PhNle. In some embodiments, XaaT5Is a residue of PhNva.
In some embodiments, XaaT1Is or includes the side chain of Ahp or Y. In some embodiments, XaaT2Is or includes the side chain of Y, W, Ahp or Bph. In some embodiments, XaaT3Is or includes the side chain of L, C or Ahp. In some embodiments, XaaT4Has a side chain ofIncluding the side chain of Bph or V. In some embodiments, XaaT5The side chain of (b) is or includes a side chain of Ahp or Bph.
In some embodiments, XaaT1Is the residue of Ahp or Y. In some embodiments, XaaT2Y, W, Ahp or Bph. In some embodiments, XaaT3Residue L, C or Ahp. In some embodiments, XaaT4Is the residue of Bph or V. In some embodiments, XaaT5Residues of Ahp or Bph.
In some embodiments, - (Xaa) y-is or includes:
-(Xaa)a1-(Xaa)a2-(Xaa)a3-(Xaa)a4-(Xaa)a5-(Xaa)a6-(Xaa)a7-(Xaa)a8-(Xaa)a9-(Xaa)a10-(Xaa)a11-(Xaa)a12-(Xaa)a13-,
wherein:
each of a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and a13 is independently 0 to 5;
(Xaa)a3is or comprises XaaT1
(Xaa)a4Is or comprises XaaT2
(Xaa)a9Is or comprises XaaT3
(Xaa)a10Is or comprises XaaT4(ii) a And is
(Xaa)a11Is or comprises XaaT5
In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and/or a13 are independently 0. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and/or a13 are independently 1. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and/or a13 are independently 2. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and/or a13 are independently 3. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and/or a13 are independently 4. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and/or a13 are independently 5. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and a13 are 1.
In some embodiments, (Xaa)a1Is or includes A. In some embodiments, a1 is 1, and (Xaa)a1Is the residue of A. Other residues may also be utilized. For example, in some embodiments, (Xaa)a1Is or includes K. In some embodiments, K is optionally linked to another moiety, such as an antibody binding moiety, via a linker. In some embodiments, (Xaa)a1Is or includes K (MePEG4c) (CH bonded to an amino group in a side chain of K3O(CH2CH2O)3CH2CH2C (O) -; see the exemplary structures as described herein). In some embodiments, a1 is 0. In some embodiments, (Xaa)a1Xaa (e.g., N-terminal residue) is linked to another Xaa (e.g., (Xaa)a13Xaa of the C-terminal residue, etc.), as described herein. For example, in some embodiments, the N-terminal residue is linked to the C-terminal cysteine (e.g., -C (o) -CH) via its amino group, through a linker, via-S-of the C-terminal cysteine2-, in which-C (O) -is bonded to an amino group, and-CH2-is bonded to-S-.
In some embodiments, (Xaa)a2Is or includes a residue whose side chain includes a heteroatom, OH or NH. In some embodiments, (Xaa)a2Is or includes a residue comprising a polar or charged side chain. Other types of residues may also be utilized, such as residues comprising hydrophobic aliphatic side chains, e.g., a. In some embodiments, (Xaa) a2Is or includes a residue whose side chain is: r, S, D, Y, A, W, K, 4Py2NH2((S) -2-amino-3- (2-aminopyridin-4-yl) propanoic acid), Cit (citrulline), F3G (3-guanidinophenylalanine), hCit (2-amino-5- (carbamoylamino) hexanoic acid; CAS: 201485-17-8), K (MePEG4c), RNdMe (N)5- [ (dimethylamino) iminomethyl]-L-ornithine; CAS: 1185841-84-2), RNMe (N5- [ imino (methylamino) methyl group]-acetate-L-ornithine; CAS: 1135616-49-7) or RNNdMe (N)5- [ (methylamino) (methylimino) methyl]-L-ornithine). In some embodiments, (Xaa)a2Is or includes a residue having a side chain of R, S, D, Y, A or W. In some embodiments, (Xaa)a2Is or includes a residue of R, S, D, Y, A, W, K, 4Py2NH2, Cit, F3G, hCit, K (MePEG4c), RNdMe, RNMe or RNNdMe. In some embodiments, (Xaa)a2Is or includes residue R, S, D, Y, A or W. In some embodiments, (Xaa)a2Is or includes R. In some embodiments, (Xaa)a2Is or includes S. In some embodiments, (Xaa)a2Is or includes D. In some embodiments, (Xaa)a2Is or includes Y. In some embodiments, (Xaa)a2Is or includes W. In some embodiments, (Xaa) a2Is or includes A. In some embodiments, (Xaa)a2Is or includes S. In some embodiments, (Xaa)a2Is or includes K. In some embodiments, (Xaa)a2Is or includes 4Py2NH 2. In some embodiments, (Xaa)a2Is or includes Cit. In some embodiments, (Xaa)a2Is or includes F3G. In some embodiments, (Xaa)a2Is or includes hCit. In some embodiments, (Xaa)a2Is or includes K (MePEG4 c). In some embodiments, (Xaa)a2Is or includes RNdMe. In some embodiments, (Xaa)a2Is or includes RNMe. In some embodiments, (Xaa)a2Is or includes RNNdMe. In some embodiments, a2 is 1.
In some embodiments, a3 is 1. In some embodiments, (Xaa)a3Is Xaa as described hereinT1
In some embodiments, a4 is 1. In some embodiments, (Xaa)a4Is Xaa as described hereinT2
In some embodiments, (Xaa)a5Is or comprises XaaT1. In some embodiments, (Xaa)a5Is or includes a residue whose side chain includes an aromatic group. In some embodiments, (Xaa)a5Is or includes a residue whose side chain includes a heteroatom, OH or NH.In some embodiments, (Xaa)a5Is or includes a residue comprising a polar or charged side chain. Other types of residues may also be utilized, such as residues comprising hydrophobic aliphatic side chains, e.g., a. In some embodiments, (Xaa) a5Is or includes a residue having a side chain of H, A, Y, S, L, W or W6N. In some embodiments, (Xaa)a5Is or includes a residue having a side chain of H, A, Y, S, L or W. In some embodiments, (Xaa)a5Is or includes residue H, A, Y, S, L, W or W6N. In some embodiments, (Xaa)a5Is or includes residue H, A, Y, S, L or W. In some embodiments, (Xaa)a5Is or includes H. In some embodiments, (Xaa)a5Is or includes A. In some embodiments, (Xaa)a5Is or includes Y. In some embodiments, (Xaa)a5Is or includes S. In some embodiments, (Xaa)a5Is or includes L. In some embodiments, (Xaa)a5Is or includes W. In some embodiments, (Xaa)a5Is or includes W6N. In some embodiments, a5 is 1.
In some embodiments, (Xaa)a6Is or includes a residue comprising a polar or charged side chain. Other types of residues may also be utilized, such as residues comprising hydrophobic aliphatic side chains, e.g., a. In some embodiments, (Xaa)a6Is or includes residues that do not include side chains. In some embodiments, (Xaa)a6Is or includes a residue having a side chain of D, R, A or Y. In some embodiments, (Xaa) a6Is or includes residue D, A, G, R or Y. In some embodiments, (Xaa)a6Is or includes D. In some embodiments, (Xaa)a6Is or includes A. In some embodiments, (Xaa)a6Is or includes G. In some embodiments, (Xaa)a6Is or includes R. In some embodiments, (Xaa)a6Is or includes Y. In some embodiments, a6 is 1.
In some embodiments, (Xaa)a7Is or includes a residue comprising a polar or charged side chain. Other types of residues may also be utilized, for example, residues comprising hydrophobic aliphatic side chainsSuch as a. In some embodiments, polar or charged amino acids may provide certain benefits, such as improved solubility for manufacturing, administration, delivery, activity, and the like. In some embodiments, (Xaa)a7Is or includes residues that do not include side chains. In some embodiments, (Xaa)a7Is or includes a residue whose side chain is that of MetO2 (methionine sulfone), D, R, A or Y. In some embodiments, (Xaa)a7Is or includes a residue having a side chain of D, R, A or Y. In some embodiments, (Xaa)a7Is or includes a residue having a side chain of D, R or S. In some embodiments, (Xaa)a7Is or includes a residue having a side chain of D, E, N or Q. In some embodiments, (Xaa) a7Is or includes a residue of MetO2, D, A, G, R, or Y. In some embodiments, (Xaa)a7Is or includes residue D, A, G, R or Y. In some embodiments, (Xaa)a7Is or includes residue D, E, N or Q. In some embodiments, (Xaa)a7Is or includes residue D, G, R or S. In some embodiments, (Xaa)a7Is or includes G. In some embodiments, (Xaa)a7Is or includes MetO 2. In some embodiments, (Xaa)a7Is or includes A. In some embodiments, (Xaa)a7Is or includes D. In some embodiments, (Xaa)a7Is or includes E. In some embodiments, (Xaa)a7Is or includes Q. In some embodiments, (Xaa)a7Is or includes N. In some embodiments, (Xaa)a7Is or includes R. In some embodiments, (Xaa)a7Is or includes S. In some embodiments, a7 is 1.
In some embodiments, (Xaa)a8Is or includes a residue comprising a hydrophobic side chain. In some embodiments, (Xaa)a8Is or includes a residue comprising an aliphatic side chain. In some embodiments, (Xaa)a8Is or includes a residue having a side chain of V, D, G, W, S, T or A. In some embodiments, (Xaa)a8Is or includes residue V, D, G, W, S, T or A. In some embodiments, (Xaa) a8Is or includes V. In some embodiments, (Xa)a)a8Is or includes D. In some embodiments, (Xaa)a8Is or includes G. In some embodiments, (Xaa)a8Is or includes W. In some embodiments, (Xaa)a8Is or includes S. In some embodiments, (Xaa)a8Is or includes T. In some embodiments, (Xaa)a8Is or includes A. In some embodiments, a8 is 1.
In some embodiments, a9 is 1. In some embodiments, (Xaa)a9Is Xaa as described hereinT3
In some embodiments, a10 is 1. In some embodiments, (Xaa)a10Is Xaa as described hereinT4
In some embodiments, a11 is 1. In some embodiments, (Xaa)a11Is Xaa as described hereinT5
In some embodiments, (Xaa)a12Is or includes a residue comprising a polar or charged side chain. In some embodiments, (Xaa)a12Is or includes residues that do not include side chains. In some embodiments, (Xaa)a12Is or includes a residue comprising a hydrophobic side chain. In some embodiments, (Xaa)a12Is or includes a residue having a side chain of D, S, G, Ahp or A. In some embodiments, (Xaa)a12Is or includes residue D, S, G, Ahp or A. In some embodiments, (Xaa)a12Is or includes D. In some embodiments, (Xaa) a12Is or includes S. In some embodiments, (Xaa)a12Is or includes G. In some embodiments, (Xaa)a12Is or includes Ahp. In some embodiments, (Xaa)a12Is or includes A. In some embodiments, a12 is 1.
In some embodiments, (Xaa)a13Is or includes a residue whose side chain includes a nucleophile. In some embodiments, (Xaa)a13Is or includes a residue whose side chain includes-S-. In some embodiments, (Xaa)a13Is or includes a residue having a side chain whose side chain is C. In some embodiments, (Xaa)a13Is or includes a residue of C. In some embodiments, a13 is 1.In some embodiments, a13 is greater than 1, and the last residue is a residue whose side chain includes a nucleophile as described herein, e.g., C. In some embodiments, (Xaa)a13Xaa (e.g., C-terminal residue) is linked to another Xaa (e.g., (Xaa)a1Xaa of the C-terminal residue, etc.), as described herein. In some embodiments, it is linked via a linker, e.g., an LT as described herein. For example, in some embodiments, the C-terminal residue is linked to the N-terminal cysteine (e.g., -C (o) -CH) via its-S-, through a linker, via the amino group of the N-terminal cysteine 2-, in which-C (O) -is bonded to an amino group, and-CH2-is bonded to-S-. In some embodiments, a residue whose side chain includes-S- (e.g., a residue of C) is linked to an amino group of another residue (e.g., -C (o) -CH) via a linker2-, in which-C (O) -is bonded to an amino group, and-CH2-is bonded to-S-.
Exemplary sequences and data thereof include those described below:
Figure BDA0003526174380000701
Figure BDA0003526174380000702
Figure BDA0003526174380000711
larger values indicate binding. And (4) performing ELISA analysis.
In some embodiments, the target binding moiety or
Figure BDA0003526174380000712
As described above and/or as utilized in the compounds in table 1.
Figure BDA0003526174380000713
Is or comprise
Figure BDA0003526174380000714
Or a salt form thereof. In some embodiments of the present invention, the,
Figure BDA0003526174380000715
is or comprise
Figure BDA0003526174380000716
Or a salt form thereof. In some embodiments of the present invention, the,
Figure BDA0003526174380000717
is or comprise
Figure BDA0003526174380000721
Or a salt form thereof. In some embodiments of the present invention, the,
Figure BDA0003526174380000722
is or comprise
Figure BDA0003526174380000723
Or a salt form thereof. In some embodiments of the present invention, the,
Figure BDA0003526174380000731
is or comprise
Figure BDA0003526174380000732
Or a salt form thereof. In some embodiments of the present invention, the,
Figure BDA0003526174380000733
is or comprise
Figure BDA0003526174380000734
Or a salt form thereof.
In some embodiments, - (Xaa) y-is or includes-XaaT6-(Xaa)y′-XaaT7-XaaT8-XaaT9-XaaT10-XaaT11-,
Wherein:
y' is 0 to 8;
XaaT6for C whose side chain is substituted1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT7is C whose side chain is optionally substituted2-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT8is a residue of proline or an amino acid analogue thereof;
XaaT9Including an optionally substituted aromatic group for its side chain or being optionally substituted C1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT10for C whose side chain is substituted1-C8A residue of an aliphatic group amino acid or an amino acid analog or a residue of an amino acid in which an amino group thereof is substituted; and is
XaaT11Including an optionally substituted aromatic group for its side chain or being optionally substituted C1-C8Aliphatic amino acids or amino acid analogues.
In some embodiments, y' is 0. In some embodiments, y' is 1. In some embodiments, y' is 2. In some embodiments, y' is 3. In some embodiments, y' is 4. In some embodiments, y' is 5. In some embodiments, y' is 6. In some embodiments, y' is 7. In some embodiments, y' is 8.
In some embodiments, XaaT6Are residues that include hydrophobic side chains. In some embodiments, XaaT6For which the side chain includes substituted C1-C8A residue of an aliphatic group. In some embodiments, the side chain is-CH2-R, wherein-CH2-is optionally substituted, and R is optionally substituted aryl or heteroaryl. In some embodiments, R is phenyl. In some embodiments, the side chain is-CH 2Ph. In some embodiments, XaaT6Is an amino acid residue, and the amino group thereof is substituted. In some embodiments, the amino group is-N (R') -. In some embodiments, R' is optionally substituted C1-C6Alkyl radical. In some embodiments, R' is methyl. In some embodiments, the side chain is that of MeF (F (-N (Me))) L, or S where a methyl group is present on the N. In some embodiments, XaaT6is-N (Me) -CH (CH)2Ph) -C (O) -. In some embodiments, XaaT6Is a residue of MeF, L or S.
In some embodiments, XaaT7Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, XaaT7Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT7Is or includes optionally substituted C3-C8An alkyl group. In some embodiments, XaaT7Is or includes optionally substituted C4-C8An alkyl group. In some embodiments, XaaT7Is or includes optionally substituted C3-C8A branched chain alkyl group. In some embodiments, XaaT7Is or includes optionally substituted C4-C8A branched chain alkyl group. In some embodiments, XaaT7The side chain of (A) is branched C3-C8An alkyl group. In some embodiments, XaaT7The side chain of (A) is branched C 4-C8An alkyl group. In some embodiments, the side chain is (CH)3)2CHCH2-. In some embodiments, the side chain is a side chain of L. In some embodiments, XaaT7Is a residue of L.
In some embodiments, XaaT8Are residues that include cyclic moieties that participate in the backbone. In some embodiments, XaaT8Is P.
In some embodiments, XaaT9Is or includes an aromatic group. In some embodiments, XaaT9Side chain of (A) is-CH2-R, wherein-CH2-is optionally substituted, and R is as described herein. In some embodiments, R is optionally substituted aryl or heteroaryl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is 4-hydroxyphenyl. In some embodiments, R is 4-phenylphenyl. In some casesIn the examples, the side chain is that of Bph. In some embodiments, XaaT9Is the residue of Bph. In some embodiments, XaaT9Including substituted C1-C8An aliphatic group. In some embodiments, the substitution is a polar or charged group, such as-OH, -COOH, or the like. In some embodiments, the side chain is a side chain of D or S. In some embodiments, XaaT9Is a residue of D or S.
In some embodiments, XaaT10Is or includes substituted C 1-C8An aliphatic group. In some embodiments, XaaT10Is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, XaaT10Is or includes optionally substituted C2-C8An alkyl group. In some embodiments, XaaT10Has a side chain of C2-C8An alkyl group. In some embodiments, XaaT10Is or includes an optionally substituted linear C2-C8An alkyl group. In some embodiments, XaaT10Has a side chain of linear C2-C8An alkyl group. In some embodiments, XaaT10Is or includes an optionally substituted branched chain C3-C8An alkyl group. In some embodiments, XaaT10The side chain of (A) is branched C3-C8An alkyl group. In some embodiments, the side chain is (CH)3)2CH-. In some embodiments, the side chain is (CH)3)2CHCH2-. In some embodiments, XaaT10Is the residue of V. In some embodiments, XaaT10Is a residue of L. In some embodiments, XaaT10Is an amino acid residue, and the amino group thereof is substituted. In some embodiments, the amino group is-N (R') -. In some embodiments, R' is optionally substituted C1-C6An alkyl group. In some embodiments, R' is methyl. In some embodiments, XaaT10Without side chains. In some embodiments, XaaT10is-N (Me) -CH2-C(O)-。
In some embodiments, Xaa T11Is or a bagIncluding aromatic groups. In some embodiments, XaaT11Side chain of (A) is-CH2-R, wherein-CH2-is optionally substituted, and R is as described herein. In some embodiments, R is optionally substituted aryl or heteroaryl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is 4-hydroxyphenyl. In some embodiments, R is 4-phenylphenyl. In some embodiments, R is optionally substituted
Figure BDA0003526174380000751
In some embodiments, R is
Figure BDA0003526174380000752
In some embodiments, the side chain is a side chain of W. In some embodiments, XaaT11Is the residue of W. In some embodiments, XaaT11Including substituted C1-C8An aliphatic group. In some embodiments, the substitution is a polar or charged group, such as-OH, -COOH, or the like. In some embodiments, the side chain is positively charged. In some embodiments, the side chain is that of R. In some embodiments, XaaT11Is the residue of R.
In some embodiments, XaaT6Is the residue of MeF. In some embodiments, XaaT7Is a residue of L. In some embodiments, XaaT8Is the residue of P. In some embodiments, XaaT9Is the residue of Bph. In some embodiments, XaaT10Is the residue of V. In some embodiments, Xaa T11Is the residue of W.
In some embodiments, - (Xaa) y-is or includes:
-(Xaa)a1-(Xaa)a2-(Xaa)a3-(Xaa)a4-(Xaa)a5-(Xaa)a6-(Xaa)a7-(Xaa)a8-(Xaa)a9-(Xaa)a10-(Xaa)a11-(Xaa)a12-,
wherein:
each of a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and a12 is independently 0 to 5;
(Xaa)a4is or comprises XaaT6
(Xaa)a6Is or comprises XaaT7
(Xaa)a7Is or comprises XaaT8
(Xaa)a8Is or comprises XaaT9
(Xaa)a9Is or comprises XaaT10(ii) a And is
(Xaa)a10Is or comprises XaaT11
In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and/or a12 are independently 0. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and/or a12 are independently 1. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and/or a12 are independently 2. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and/or a12 are independently 3. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and/or a12 are independently 4. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and/or a12 are independently 5. In some embodiments, a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and a12 are 1.
In some embodiments, (Xaa)a1Is or includes A. In some embodiments, a1 is 1, and (Xaa) a1Is the residue of A. In some embodiments, a1 is 0. In some embodiments, (Xaa)a1Xaa (e.g., N-terminal residue) is linked to another Xaa (e.g., (Xaa)a13Xaa of the C-terminal residue, etc.), as described herein. For example, in some embodiments, the N-terminal residue is linked to the C-terminal cysteine (e.g., -C (o) -CH) via its amino group, through a linker, via-S-of the C-terminal cysteine2-, in which-C (O) -is bonded to an amino group, and-CH2-is bonded to-S-.
In some embodiments, (Xaa)a2Is or comprises Xaa comprising a hydrophobic side chainHAnd (c) a residue. In some embodiments, the side chain is-CH3. In some embodiments, the side chain is (CH)3)2CHCH2-. In some embodiments, Xaa is a residue of L. In some embodiments, Xaa is a residue of a. In some embodiments, Xaa is a residue of P. In some embodiments, (Xaa)a2Is or includes L. In some embodiments, (Xaa)a2Is or includes A. In some embodiments, (Xaa)a2Is or includes P. In some embodiments, a2 is 1.
In some embodiments, XaaHThe side chain of (A) includes substituted C1-C8An aliphatic group. In some embodiments, the side chain is or includes optionally substituted C2-C8An aliphatic group. In some embodiments, the side chain is or includes optionally substituted C 2-C8An alkyl group. In some embodiments, the side chain is or includes optionally substituted C3-C8An alkyl group. In some embodiments, the side chain is or includes optionally substituted C4-C8An alkyl group. In some embodiments, the side chain is or includes optionally substituted C3-C8A branched chain alkyl group. In some embodiments, the side chain is or includes optionally substituted C4-C8A branched chain alkyl group. In some embodiments, the side chain is branched C3-C8An alkyl group. In some embodiments, the side chain is branched C4-C8An alkyl group. In some embodiments, the side chain is methyl. In some embodiments, the side chain is (CH)3)2CHCH2-. In some embodiments, XaaHIs a residue of L. In some embodiments, XaaHIs the residue of A.
In some embodiments, (Xaa)a3Is or includes a residue comprising a basic side chain (positively charged side chain). In some embodiments, the side chain of Xaa includes an optionally substituted aromatic basic moiety. In some embodiments, the side chain includes an optionally substituted imidazolyl group. In some embodiments, the side chain of Xaa includes an optionally substituted non-aromatic basic moiety. In some embodiments, the side chain comprises an optionally substituted guanidino group. In some embodiments, the side chain comprisesA substituted amino group is selected. Other types of residues may also be utilized, such as residues comprising hydrophobic aliphatic side chains, e.g., a. In some embodiments, the side chain is a side chain of H. In some embodiments, the side chain is that of R. In some embodiments, the side chain is that of a. In some embodiments, Xaa is a residue of H. In some embodiments, Xaa is the residue of R. In some embodiments, (Xaa) a3Is or includes Xaa as described hereinHAnd (c) a residue. In some embodiments, Xaa is a residue of a. In some embodiments, (Xaa)a3Is or includes H. In some embodiments, (Xaa)a3Is or includes R. In some embodiments, (Xaa)a3Is or includes A. In some embodiments, a3 is 1.
In some embodiments, (Xaa)a4Is or comprises XaaT6. In some embodiments, a4 is 1. In some embodiments, (Xaa)a4Is Xaa as described hereinT6. In some embodiments, (Xaa)a4Is a residue of MeF or L. In some embodiments, (Xaa)a4Is or includes MeF. In some embodiments, (Xaa)a4Is or includes L. In some embodiments, a4 is 1.
In some embodiments, (Xaa)a5Is or comprises XaaH. In some embodiments, (Xaa)a5Is or includes C whose side chain is or includes optionally substituted1-C8Xaa of an aliphatic group. In some embodiments, the side chain is methyl. In some embodiments, Xaa is Xaa as described hereinT10. In some embodiments, Xaa is the residue of V. In some embodiments, Xaa is a residue of a. In some embodiments, Xaa is a residue of MeG (methyl on amino). In some embodiments, (Xaa)a5Is or includes V. In some embodiments, (Xaa) a5Is or includes A. In some embodiments, (Xaa)a5Is or includes MeG. In some embodiments, a5 is 1.
In some embodiments, (Xaa)a6As described herein (Xaa)a2. In some embodiments, (Xaa)a6Is or comprise a hydrophobic sideXaa of the chainHAnd (c) a residue. In some embodiments, the side chain is-CH3. In some embodiments, the side chain is (CH)3)2CHCH2-. In some embodiments, Xaa is a residue of L. In some embodiments, Xaa is a residue of a. In some embodiments, Xaa is a residue of P. In some embodiments, (Xaa)a6Is or includes L. In some embodiments, (Xaa)a6Is or includes A. In some embodiments, (Xaa)a6Is or includes P. In some embodiments, a6 is 1.
In some embodiments, (Xaa)a6Is or comprises XaaT7. In some embodiments, a6 is 1. In some embodiments, (Xaa)a6Is Xaa as described hereinT7. In some embodiments, (Xaa)a6Is a residue of L or P.
In some embodiments, (Xaa)a7Is or comprises XaaT8. In some embodiments, a7 is 1. In some embodiments, (Xaa)a7Is Xaa as described hereinT8. In some embodiments, (Xaa)a7Is the residue of P.
In some embodiments, (Xaa) a8Is or comprises XaaT9. In some embodiments, a8 is 1. In some embodiments, (Xaa)a8Is Xaa as described hereinT9. In some embodiments, (Xaa)a8Is the residue of Bph. In some embodiments, (Xaa)a8Is a residue of D or S.
In some embodiments, (Xaa)a9Is or comprises XaaT10. In some embodiments, a9 is 1. In some embodiments, (Xaa)a9Is Xaa as described hereinT10. In some embodiments, (Xaa)a9V, L or MeG.
In some embodiments, (Xaa)a10Is or comprises XaaT11. In some embodiments, a10 is 1. In some embodiments, (Xaa)a10Is Xaa as described hereinT11. In some embodiments, (Xaa)a10Is a residue of W or R.
In some embodiments, (Xaa)a11Is or comprises XaaH. In some embodiments, (Xaa)a11Is or includes C whose side chain is or includes optionally substituted1-C8Xaa of an aliphatic group. In some embodiments, the side chain is methyl. In some embodiments, the side chain is isopropyl. In some embodiments, Xaa is Xaa as described hereinT10. In some embodiments, Xaa is the residue of V. In some embodiments, Xaa is the residue of V. In some embodiments, Xaa is a residue of a. In some embodiments, Xaa is a residue of MeG (methyl on amino). In some embodiments, (Xaa) a11Is or includes V. In some embodiments, (Xaa)a11Is or includes A. In some embodiments, (Xaa)a11Is or includes MeG. In some embodiments, a11 is 1.
In some embodiments, (Xaa)a12Is or includes a residue whose side chain includes a nucleophile. In some embodiments, (Xaa)a12Is or includes a residue whose side chain includes-S-. In some embodiments, (Xaa)a12Is or includes a residue having a side chain whose side chain is C. In some embodiments, (Xaa)a12Is or includes a residue of C. In some embodiments, a12 is 1. In some embodiments, a12 is greater than 1, and the last residue is a residue whose side chain includes a nucleophile as described herein, e.g., C. In some embodiments, (Xaa)a12Xaa (e.g., C-terminal residue) is linked to another Xaa (e.g., (Xaa)a1Xaa of the C-terminal residue, etc.), as described herein. For example, in some embodiments, the C-terminal residue is linked to the N-terminal cysteine (e.g., -C (o) -CH) via its-S-, through a linker, via the amino group of the N-terminal cysteine2-, in which-C (O) -is bonded to an amino group, and-CH2-is bonded to-S-. In some embodiments, a residue whose side chain includes-S- (e.g., a residue of C) is linked to an amino group of another residue (e.g., -C (o) -CH) via a linker 2-, in which-C (O) -is bonded to an amino group, and-CH2-is bonded to-S-.
Exemplary sequences and data thereof include those described below:
Figure BDA0003526174380000781
Figure BDA0003526174380000791
Figure DA00035261743831087499
larger values indicate binding. And (4) performing ELISA analysis.
In some embodiments, the target binding moiety or
Figure BDA0003526174380000792
As described above and/or as utilized in the compounds in table 1. In some embodiments of the present invention, the,
Figure BDA0003526174380000793
is or comprise
Figure BDA0003526174380000801
Or a salt form thereof. In some embodiments of the present invention, the,
Figure BDA0003526174380000802
is or comprise
Figure BDA0003526174380000803
Or a salt form thereof.
In some embodiments, the target binding moiety or
Figure BDA0003526174380000804
Or- (Xaa) y-is or includes the following peptide:
(1) a polypeptide having an amino acid sequence represented by any one of SEQ ID nos. 1 to 34:
Figure BDA0003526174380000805
Figure BDA0003526174380000811
(2) a polypeptide having an amino acid sequence represented by any one of SEQ ID nos. 1 to 34, wherein the amino acid residue at the N-terminus is chloroacetylated (e.g., at the amino group thereof);
(3) a polypeptide having an amino acid sequence with a deletion, addition, substitution or insertion of one or more amino acids in any one of SEQ ID nos. 1-34, which does not include a deleted amino acid sequence having a Cys at the C-terminus in SEQ ID nos. 1-34;
(4) a polypeptide having an amino acid sequence represented by any one of SEQ ID nos. 1-34 having a deletion, addition, substitution, or insertion of one or more amino acids in any one of SEQ ID nos. 1-34, excluding a deleted amino acid sequence having a Cys at the C-terminus in any one of SEQ ID nos. 1-34, wherein the amino acid at the N-terminus is chloroacetylated (e.g., at its amino acid); or
(5) The polypeptide according to any one of (1) to (4) above, wherein the polypeptide has a cyclized structure.
In some embodiments, the target binding moiety or
Figure BDA0003526174380000821
Or- (Xaa) y-is or includes the following peptide:
(1) a polypeptide having an amino acid sequence represented by SEQ ID No.1 or 2:
Ala Arg Ahp Tyr His Asp Gly Val Leu Bph Ahp Asp Cys(SEQ ID NO.1),
Ala Leu His MePhe Val Leu Pro Bph Val Trp Val Cys(SEQ ID NO.2);
(2) a polypeptide having an amino acid sequence represented by SEQ ID No.1 or 2, wherein Ala at the N-terminus is chloroacetylated Ala;
(3) a polypeptide having a deletion, addition, substitution, or insertion of one or more amino acids in SEQ ID No.1 or 2 in the amino acid sequence, which does not include an amino acid sequence having a deletion of Cys at the C-terminus in SEQ ID No.1 or 2;
(4) a polypeptide having an amino acid sequence represented by SEQ ID No.1 or 2, wherein Ala at the N-terminus is chloroacetylated Ala, having a deletion, addition, substitution, or insertion of one or more amino acids in SEQ ID No.1 or 2, which excludes a deleted amino acid sequence having Cys at the C-terminus in SEQ ID No.1 or 2; or
(5) The polypeptide according to any one of (1) to (4) above, wherein the polypeptide has a cyclized structure.
In some embodiments, amino acid residues, e.g., at the N-terminus, such as Ala via-C (O) -CH 2-is linked to Cys, wherein-C (O) -is bonded to Ala, and-CH2-S-bonded to Cys. In some embodiments, the N-terminal amino acid residue (e.g., Ala) is attached by reacting a chloroacetylated amino acid residue (e.g., Ala) with-SH from Cys under suitable conditions.
In some embodiments, the amino acid substitution is a conservative substitution. In some embodiments, the substitution does not significantly affect the structure, properties, and/or activity of the peptide and/or protein. In some embodiments, examples of groups of amino acids whose side chains have similar chemical properties include 1) aliphatic side chains: glycine, alanine, valine, leucine, and isoleucine; 2) aliphatic hydroxyl side chain: serine and threonine; 3) amide-containing side chains: asparagine and glutamine; 4) aromatic side chain: phenylalanine, tyrosine and tryptophan; 5) basic side chain: lysine, arginine and histidine; 6) acidic side chain: aspartic acid and glutamic acid; and 7) sulfur containing side chains: cysteine and methionine. In some embodiments, the conservative amino acid substitution is selected from valine-leucine-isoleucine, phenylalanine-tyrosine-tryptophan, lysine-arginine, alanine-valine, glutamic acid-aspartic acid, and asparagine-glutamine. The aforementioned amino acids may be proteinaceous or non-proteinaceous amino acids. Those skilled in the art will appreciate that, depending on the situation, amino acids may be grouped in other ways that are appropriate for the intended purpose based on structure, properties, activity, etc. In some embodiments, the present disclosure provides a target binding moiety that is or comprises an amino acid sequence having a deletion, substitution, insertion and/or addition of 1 to 5 amino acids, preferably 4 or less, 3 or less, 2 or less, more preferably one amino acid or less in the amino acid sequence represented by one of SEQ ID nos. 1 to 34 and that can bind to CD 38.
In some embodiments, the target binding moiety or
Figure BDA0003526174380000831
Is or includes a sequence represented by one of SEQ ID NO.1 to 34. In some embodiments, the target binding moiety is or comprises a cyclic structure.
In some embodiments, the target binding moiety is derived from, or
Figure BDA0003526174380000832
The following are: a structure selected from the following S-1 to S-32 (wherein SEQ ID No. of amino acid sequence is indicated) or a pharmaceutically acceptable salt thereof:
Figure BDA0003526174380000841
Figure BDA0003526174380000851
Figure BDA0003526174380000861
Figure BDA0003526174380000871
Figure BDA0003526174380000881
Figure BDA0003526174380000891
Figure BDA0003526174380000901
Figure BDA0003526174380000911
Figure BDA0003526174380000921
Figure BDA0003526174380000931
Figure BDA0003526174380000941
Figure BDA0003526174380000951
Figure BDA0003526174380000961
Figure BDA0003526174380000971
Figure BDA0003526174380000981
Figure BDA0003526174380000991
Figure BDA0003526174380001001
Figure BDA0003526174380001011
Figure BDA0003526174380001021
Figure BDA0003526174380001031
Figure BDA0003526174380001041
Figure BDA0003526174380001051
Figure BDA0003526174380001061
Figure BDA0003526174380001071
Figure BDA0003526174380001081
Figure BDA0003526174380001091
Figure BDA0003526174380001101
Figure BDA0003526174380001111
in some embodiments, the target binding moiety is derived from, or
Figure BDA0003526174380001112
The following are: a structure selected from the following S-33 to S-39 (wherein SEQ ID No. of amino acid sequence is indicated) or a pharmaceutically acceptable salt thereof:
Figure BDA0003526174380001113
Figure BDA0003526174380001121
Figure BDA0003526174380001131
Figure BDA0003526174380001141
in addition, various structures (e.g., S-1 to S-39) were assessed in various assays and shown to bind to CD 38.
As will be appreciated by those of skill in the art, structures (e.g., S-1 to S-39) can be attached to the remainder of the molecule (e.g., optionally via the antibody-binding portion of a linker) via a variety of suitable means in accordance with the present disclosure (e.g., via side chains (e.g., amino groups of certain side chains), N-termini, C-termini, etc.).
In some embodiments, the peptide unit, for example, has
Figure BDA0003526174380001142
Target binding moieties of structures orSalts thereof include functional groups in an amino acid residue that are reactive with functional groups of another amino acid residue. In some embodiments, a peptide unit comprises an amino acid residue having a side chain that includes a functional group that can react with another functional group of a side chain of another amino acid residue to form a bond (e.g., see moieties in table a-1, table 1, etc.). In some embodiments, one functional group of one amino acid residue is linked to a functional group of another amino acid residue to form a bond (or bridge). The bond is to the backbone atom of the peptide unit and does not include the backbone atom. In some embodiments, a peptide unit comprises a bond formed by two side chains of non-adjacent amino acid residues. In some embodiments, the bond is bonded to two backbone atoms of two non-adjacent amino acid residues. In some embodiments, both backbone atoms bonded to the bond are carbon atoms. In some embodiments, the key has L bStructure of, wherein LbIs L as described in this disclosureaWherein L isaNot a covalent bond. In some embodiments, Laincluding-Cy-. In some embodiments, Laincluding-Cy-wherein-Cy-is optionally substituted heteroaryl. In some embodiments, -Cy-is
Figure BDA0003526174380001151
In some embodiments, LaIs composed of
Figure BDA0003526174380001152
In some embodiments, such LaCan be derived from the side chain of an amino acid residue3Formation of a group and a side chain of another amino acid residue. In some embodiments, the bond is formed via the attachment of two thiol groups, e.g., two cysteine residues. In some embodiments, Laincluding-S-S-. In some embodiments, Lais-CH2-S-S-CH2-. In some embodiments, the bond is via an amino group (e.g., -NH in the side chain of a lysine residue)2) And a carboxylic acid group (e.g., -COOH in the side chain of an aspartic acid or glutamic acid residue). In some embodiments, Lacomprising-C (O) -N (R') -. In some embodiments,Laincluding-C (O) -NH-. In some embodiments, Lais-CH2CONH-(CH2)3-. In some embodiments, Laincluding-C (O) -N (R ') -, where R' is R, and forms a ring with the R group on the peptide backbone (e.g., as in A-34). In some embodiments, LaIs- (CH)2)2-N(R′)-CO--(CH2)2-. In some embodiments, -Cy-is optionally substituted phenylene. In some embodiments, -Cy-is optionally substituted 1, 2-phenylene. In some embodiments, L aIs composed of
Figure BDA0003526174380001153
In some embodiments, LaIs composed of
Figure BDA0003526174380001154
In some embodiments, LaIs optionally substituted divalent C2-20A divalent aliphatic group. In some embodiments, LaIs optionally substituted- (CH)2)9-CH=CH-(CH2)9-. In some embodiments, LaIs- (CH)2)3-CH=CH-(CH2)3-。
In some embodiments, the two amino acid residues bonded to the bond are separated by 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or more than 15 amino acid residues (not including the two amino acid residues bonded to the bond). In some embodiments, the number is 1. In some embodiments, the number is 2. In some embodiments, the number is 3. In some embodiments, the number is 4. In some embodiments, the number is 5. In some embodiments, the number is 6. In some embodiments, the number is 7. In some embodiments, the number is 8. In some embodiments, the number is 9. In some embodiments, the number is 10. In some embodiments, the number is 11. In some embodiments, the number is 12. In some embodiments, the number is 13. In some embodiments, the number is 14. In some embodiments, the number is 15.
In some casesIn embodiments, the target binding moiety comprises a peptide unit, and the antibody binding moiety is attached to a backbone atom of the peptide unit, optionally via a linker. In some embodiments, the target binding moiety comprises a peptide unit, and the antibody binding moiety is attached to an atom of a side chain of an amino acid residue of the peptide unit, optionally via a linker, e.g. via an atom or group in the side chain. For example, in some embodiments, the antibody-binding moiety is via the side chain-SH, -OH, -COOH, or-NH 2And (4) connecting.
Amino acids
In some embodiments, provided compounds and agents may comprise one or more amino acid moieties, for example in a universal antibody binding moiety, a linker moiety, and the like. The amino acid moiety can be a natural amino acid or an amino acid moiety of an unnatural amino acid. In some embodiments, the amino acid has the structure of formula a-I:
NH(Ra1)-La1-C(Ra2)(Ra3)-La2-COOH,
A-I
or a salt thereof, wherein each variable is independently as described in the disclosure. In some embodiments, for example, amino acid residues of amino acids having the structure of formulas A-I have-N (R)a1)-La1-C(Ra2)(Ra3)-La2-CO-structure. In some embodiments, each amino acid residue in the peptide independently has-N (R)a1)-La1-C(Ra2)(Ra3)-La2-CO-structure.
In some embodiments, La1Is a covalent bond. In some embodiments, the compounds of formula A-I have NH (R)a1)-C(Ra2)(Ra3)-La2-COOH structure. In some embodiments, La2is-CH2SCH2-。
In some embodiments, La2Is a covalent bond. In some embodiments, the compounds of formula A-I have NH (R)a1)-La1-C(Ra2)(Ra3) -COOH structure. In some embodiments, the amino acid residue has-N (R)a1)-La1-C(Ra2)(Ra3) -CO-structure. In some embodiments, La1is-CH2CH2S-. In some embodiments, La1is-CH2CH2S-, in which CH2Bonded to NH (R)a1)。
In some embodiments, La1Is a covalent bond and La2Is a covalent bond. In some embodiments, the compounds of formula A-I have NH (R) a1)-C(Ra2)(Ra3) -COOH structure. In some embodiments, the compounds of formula A-I have NH (R)a1)-CH(Ra2) -COOH structure. In some embodiments, the compounds of formula A-I have NH (R)a1)-CH(Ra3) -COOH structure. In some embodiments, the compounds of formula a-I have NH2-CH(Ra2) -COOH structure. In some embodiments, the compounds of formula a-I have NH2-CH(Ra3) -COOH structure. In some embodiments, the amino acid residue has-N (R)a1)-C(Ra2)(Ra3) -CO-structure. In some embodiments, the amino acid residue has-N (R)a1)-CH(Ra2) -CO-structure. In some embodiments, the amino acid residue has-N (R)a1)-CH(Ra3) -CO-structure. In some embodiments, the amino acid residue has an-NH-CH (R)a2) -CO-structure. In some embodiments, the amino acid residue has an-NH-CH (R)a3) -CO-structure.
In some embodiments, LaIs a covalent bond. In some embodiments, LaIs optionally substituted C1-6A divalent aliphatic group. In some embodiments, LaIs optionally substituted C1-6An alkylene group. In some embodiments, Lais-CH2-. In some embodiments, Lais-CH2CH2-. In some embodiments, Lais-CH2CH2CH2-。
In some embodiments, R' is R. In some embodiments, Ra1Is R, wherein R is as described in the disclosure. In some embodiments, Ra1Is R, wherein R is methyl. In some embodiments, R a2Is R, wherein R is as described in the disclosure. In some embodiments, Ra3Is R, wherein R is as described in the disclosure. In some embodiments, Ra1、Ra2And Ra3Each of which is independently R, wherein R is as described in the disclosure.
In some embodiments, Ra1Is hydrogen. In some embodiments, Ra2Is hydrogen. In some embodiments, Ra3Is hydrogen. In some embodiments, Ra1Is hydrogen, and Ra2And Ra3At least one of which is hydrogen. In some embodiments, Ra1Is hydrogen, Ra2And Ra3One of which is hydrogen and the other is not hydrogen. In some embodiments, Ra2is-La-R and Ra3is-H. In some embodiments, Ra3is-La-R and Ra2is-H. In some embodiments, Ra2is-CH2-R and Ra3is-H. In some embodiments, Ra3is-CH2-R and Ra2is-H. In some embodiments, Ra2Is R and Ra3is-H. In some embodiments, Ra3Is R and Ra2is-H.
In some embodiments, Ra2is-La-R, wherein R is as described in the disclosure. In some embodiments, Ra2is-La-R, wherein R is an optionally substituted group selected from: c3-30Cycloaliphatic radical, C5-30An aryl group, a 5-to 30-membered heteroaryl group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon, and a 3-to 30-membered heterocyclyl group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, R a2is-La-R, wherein R is an optionally substituted group selected from: c6-30Aryl, and 5 to 30 membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, Ra2Is the side chain of an amino acid. In some embodiments, Ra2Are the side chains of standard amino acids.
In some embodiments of the present invention, the,Ra3is-La-R, wherein R is as described in the disclosure. In some embodiments, Ra3is-La-R, wherein R is an optionally substituted group selected from: c3-30Cycloaliphatic radical, C5-30An aryl group, a 5-to 30-membered heteroaryl group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon, and a 3-to 30-membered heterocyclyl group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, Ra3is-La-R, wherein R is an optionally substituted group selected from: c6-30Aryl, and 5 to 30 membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, Ra3Is the side chain of an amino acid. In some embodiments, Ra3Are the side chains of standard amino acids.
In some embodiments, R is optionally substituted C1-6An aliphatic group. In some embodiments, R is optionally substituted C 1-6An alkyl group. In some embodiments, R is-CH3. In some embodiments, R is optionally substituted pentyl. In some embodiments, R is n-pentyl.
In some embodiments, R is a cyclic group. In some embodiments, R is optionally substituted C3-30A cycloaliphatic radical. In some embodiments, R is cyclopropyl.
In some embodiments, R is an optionally substituted aromatic group and the amino acid residue of the amino acid of formula A-I is XaaA. In some embodiments, Ra2Or Ra3is-CH2-R, wherein R is optionally substituted aryl or heteroaryl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is phenyl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is 4-trifluoromethylphenyl. In some embodiments, R is 4-phenylphenyl. In some embodiments, R is an optionally substituted 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, R is a group having 1 to 5 substituents independently selected from oxygen,Optionally substituted 5-to 14-membered heteroaryl of nitrogen and sulfur heteroatoms. In some embodiments, R is
Figure BDA0003526174380001181
In some embodiments, R is optionally substituted pyridinyl. In some embodiments, R is 1-pyridyl. In some embodiments, R is 2-pyridyl. In some embodiments, R is 3-pyridyl. In some embodiments, R is
Figure BDA0003526174380001182
In some embodiments, R' is — COOH. In some embodiments, the compound of an amino acid residue of an amino acid of formula a-I is XaaN
In some embodiments, R' is-NH2. In some embodiments, the compound of an amino acid residue of an amino acid of formula a-I is XaaP
In some embodiments, Ra2Or Ra3Is R, wherein R is C as described in the disclosure1-20An aliphatic group. In some embodiments, the compound of an amino acid residue of an amino acid of formula a-I is XaaH. In some embodiments, R is-CH3. In some embodiments, R is ethyl. In some embodiments, R is propyl. In some embodiments, R is n-propyl. In some embodiments, R is butyl. In some embodiments, R is n-butyl. In some embodiments, R is pentyl. In some embodiments, R is n-pentyl. In some embodiments, R is cyclopropyl.
In some embodiments, Ra1、Ra2And Ra3Two or more of which are R, and together form an optionally substituted ring as described in the disclosure.
In some embodiments, Ra1And Ra2And Ra3Are R and together form a radical other than Ra1An optionally substituted 3-to 6-membered ring having no additional ring heteroatoms beyond the nitrogen atom to which it is bonded. In some embodiments, the ring formed is a 5-membered ring, such as prolineIn (1).
In some embodiments, Ra2And Ra3Are R, and together form an optionally substituted 3-to 6-membered ring as described in the disclosure. In some embodiments, Ra2And Ra3Are R, and together form an optionally substituted 3-to 6-membered ring having one or more nitrogen ring atoms. In some embodiments, Ra2And Ra3Are R and together form an optionally substituted 3-to 6-membered ring having one and not more than one ring heteroatom which is a nitrogen atom. In some embodiments, the ring is a saturated ring.
In some embodiments, the amino acid is a natural amino acid. In some embodiments, the amino acid is an unnatural amino acid. In some embodiments, the amino acid is an alpha-amino acid. In some embodiments, the amino acid is a β -amino acid. In some embodiments, the compounds of formula a-I are natural amino acids. In some embodiments, the compounds of formula a-I are unnatural amino acids.
In some embodiments, the amino acid comprises a hydrophobic side chain. In some embodiments, the amino acid having a hydrophobic side chain is A, V, I, L, M, F, Y or W. In some embodiments, the amino acid with a hydrophobic side chain is A, V, I, L, M or F. In some embodiments, the amino acid with a hydrophobic side chain is A, V, I, L or M. In some embodiments, the amino acid with a hydrophobic side chain is A, V, I or L. In some embodiments, the hydrophobic side chain is R, wherein R is C 1-10An aliphatic group. In some embodiments, R is C1-10An alkyl group. In some embodiments, R is methyl. In some embodiments, R is ethyl. In some embodiments, R is propyl. In some embodiments, R is butyl. In some embodiments, R is pentyl. In some embodiments, R is n-pentyl. In some embodiments, the amino acid with a hydrophobic side chain is NH2CH(CH2CH2CH2CH2CH3) COOH. In some embodiments, the amino acid having a hydrophobic side chain is (S) -NH2CH(CH2CH2CH2CH2CH3) COOH. In some embodiments, hydrophobicThe amino acid of the side chain is (R) -NH2CH(CH2CH2CH2CH2CH3) COOH. In some embodiments, the hydrophobic side chain is-CH2R, wherein R is optionally substituted phenyl. In some embodiments, R is phenyl. In some embodiments, R is phenyl substituted with one or more hydrocarbyl groups. In some embodiments, R is 4-phenylphenyl. In some embodiments, the amino acid with a hydrophobic side chain is NH2CH(CH2-4-phenylphenyl) COOH. In some embodiments, the amino acid having a hydrophobic side chain is (S) -NH2CH(CH2-4-phenylphenyl) COOH. In some embodiments, the amino acid having a hydrophobic side chain is (R) -NH2CH(CH2-4-phenylphenyl) COOH.
In some embodiments, the amino acid comprises a positively charged side chain as described herein (e.g., at physiological pH). In some embodiments, such amino acids include a basic nitrogen in the side chain. In some embodiments, such amino acid is Arg, His, or Lys. In some embodiments, such amino acid is Arg. In some embodiments, such amino acid is His. In some embodiments, such an amino acid is Lys.
In some embodiments, the amino acid comprises a negatively charged side chain as described herein (e.g., at physiological pH). In some embodiments, such amino acids include-COOH in the side chain. In some embodiments, such amino acid is Asp. In some embodiments, such amino acid is Glu.
In some embodiments, the amino acid comprises a side chain comprising an aromatic group as described herein. In some embodiments, such amino acid is Phe, Tyr, Trp, or His. In some embodiments, such amino acid is Phe. In some embodiments, such amino acid is Tyr. In some embodiments, such amino acid is Trp. In some embodiments, such amino acid is His. In some embodiments, such amino acid is NH2-CH(CH2-4-phenylphenyl) -COOH. In some embodiments, such amino acids are (S) -NH2-CH(CH2-4-phenylphenyl) -COOH. In some embodiments, such amino groupsThe acid being (R) -NH2-CH(CH2-4-phenylphenyl) -COOH.
In some embodiments, the amino acid is a known proteinogenic amino acid that is naturally encoded or found in the genetic code of any organism, or a non-proteinogenic amino acid that is not naturally encoded or found in the genetic code of any organism. Examples of non-proteinogenic amino acids include alpha, alpha-disubstituted amino acids (alpha-methylalanine, etc.), N-alkyl-alpha-amino acids, and N-alkyl-alpha-D-amino acids, and the backbone structure may differ from that of the natural type of amino acid. Examples of such amino acids include β -amino acids and amino acids having a side chain structure different from that of the natural type (e.g., norleucine, homohistidine and hydroxyproline).
In some embodiments, the amino acid is selected from:
bph beta- (4-biphenylyl) -alanine
MetO2 methionine sulfone
Hse homoserine
Har N6-carbamimidoyl-L-lysine
da D-alanine
W6N (S) -2-amino-3- (1H-pyrrolo [2, 3-c ] pyridin-3-yl) propionic acid
Ano (S) -2-aminononanoic acid
Ado (S) -2-aminoundecanoic acid
PhNle (S) -2-amino-6-phenylhexanoic acid
PhNva (S) -2-amino-5-phenylpentanoic acid
Nal2 (2-naphthyl) alanine
Cit citrulline
F3G 3-guanidinophenylalanine
RNMe N5- [ imino (methylamino) methyl group]-acetate-L-ornithine; CAS: 1135616-49-7
RNNdMe N5- [ (methylamino) (methylimino) methyl]-L-ornithine
RNdMe N5- [ (dimethylamino) imino groupMethyl radical]-L-ornithine; CAS: 1185841-84-2
hCit 2-amino-5- (carbamoylamino) hexanoic acid; CAS: 201485-17-8
4Py2NH2 (S) -2-amino-3- (2-aminopyridin-4-yl) propionic acid
Target
In some embodiments, the present disclosure provides techniques for selectively directing an agent comprising a target binding moiety (e.g., an ARM compound), an antibody, and an immune cell (e.g., an NK cell) to a desired target site comprising one or more targets. As will be appreciated by those skilled in the art, the provided techniques are applicable to various types of targets, including, in particular, targets of CD 38.
In some embodiments, the target is damaged or defective tissue. In some embodiments, the target is damaged tissue. In some embodiments, the target is a defective tissue. In some embodiments, the target is associated with a disease, disorder, or condition, such as cancer, a wound, and the like. In some embodiments, the target is a tumor. In some embodiments, the target is or comprises a diseased cell. In some embodiments, the target is or comprises a cancer cell. In some embodiments, the target is a foreign object. In some embodiments, the target is or comprises an infectious agent. In some embodiments, the target is a microorganism. In some embodiments, the target is or comprises a bacterium. In some embodiments, the target is or comprises a virus. In some embodiments, the target comprises or expresses CD 38.
In many embodiments, the target is a tissue and/or cell associated with a disease, disorder, or condition, in particular various types of cancer. In some embodiments, the target is or includes a cell associated with a condition, disorder, or disease. In some embodiments, the target is or comprises a cell associated with cancer. In some embodiments, the cell comprises or expresses CD 38. Furthermore, the present disclosure provides techniques that are particularly useful for selectively targeting cancer cells that include or express CD38 by the immune system via, for example, recruitment of antibodies (e.g., endogenous antibodies) and immune cells through the use of ARM.
The target site typically includes one or more physical, chemical, and/or biomarker, such as CD38, which may be utilized, for example, by a target binding portion of a provided compound (e.g., ARM) to selectively recruit antibodies and/or fragments thereof and/or immune cells to the target.
In some embodiments, the cells of the target site include one or more characteristic agents suitable for targeting CD38, for example. In some embodiments, such agents are proteins and/or fragments thereof. In some embodiments, such agents are antigens associated with a disease, disorder, or condition. In some embodiments, the target site and/or cells thereof comprise and/or express CD38, and the target binding moiety of the provided ARM can bind to CD 38.
Connector section
In some embodiments, the antibody binding moiety is optionally linked to the target binding moiety via a linker moiety. Various types and/or for various purposes of linker moieties may be utilized in accordance with the present disclosure, such as for antibody-drug conjugates and the like.
The linker moiety may be divalent or multivalent. In some embodiments, the linker moiety is divalent. In some embodiments, the linker is multivalent and connects more than two moieties.
In some embodiments, the linker moiety is L. In some embodiments, L is a covalent bond, or a divalent or multivalent optionally substituted straight or branched chain C comprising one or more aliphatic groups, aryl groups, heteroaliphatic groups having 1 to 20 heteroatoms, heteroaromatic groups having 1 to 20 heteroatoms, or any combination thereof1-100A group wherein one or more methylene units of said group are optionally and independently replaced by: c1-6Alkylene radical, C1-6Alkenylene, divalent C with 1 to 5 heteroatoms1-6Heteroaliphatic, -C.ident.C-, -Cy-, -C (R')2-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-C(O)C(R′)2N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R ') -, -C (O) S-, -C (O) O-, -P (O) -, -P (O) (SR') -, -P (O) (R ') -, -P (O) (NR') -, -P (S) (OR ') -, -P (S) (SR') -, -P (S) (R ') -, -P (S) (NR') -, -P (S ') -, -P (R') -, -P (OR ') -, -P (SR') -, -P (NR ') -, an amino acid residue OR- [ (-O-C (R'))2-C(R′)2-)n]-, where n is 1 to 20. In some embodiments, each amino acid residue is independently a residue of an amino acid having the structure of formula a-I or a salt thereof. In some embodiments, each amino acid residue independently has-N (R)a1)-La1-C(Ra2)(Ra3)-La2-CO-or a salt form thereof.
In some embodiments, L is divalent. In some embodiments, L is selected from C 1-100Aliphatic radical and C having 1 to 50 heteroatoms1-100A divalent or optionally substituted straight or branched chain radical of a heteroaliphatic group, wherein one or more methylene units of said radical are optionally and independently replaced by: c1-6Alkylene radical, C1-6Alkenylene, divalent C with 1 to 5 heteroatoms1-6Heteroaliphatic, -C.ident.C-, -Cy-, -C (R')2-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-C(O)C(R′)2N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R ') -, -C (O) S-, -C (O) O-, -P (O) -, -P (O) (SR') -, -P (O) (R ') -, -P (O) (NR') -, -P (S) (OR ') -, -P (S) (SR') -, -P (S) (R ') -, -P (S) (NR') -, -P (S ') -, -P (R') -, -P (OR ') -, -P (SR') -, -P (NR ') -, amino acid OR- [ (-O-C (R'))2-C(R′)2-)n]-。
In some embodiments, L is a covalent bond. In some embodiments, L is a divalent optionally substituted straight or branched chain C1-100An aliphatic group wherein one or more methylene units of said group are optionally and independently replaced. In some embodiments, L is a divalent optionally substituted straight or branched chain C6-100Arylaliphatic group, wherein one or more methylene units of the group are optionally and independently replaced. In some implementationsIn one embodiment, L is a divalent optionally substituted straight or branched chain C having 1 to 20 heteroatoms 5-100Heteroarylaliphatic group, wherein one or more methylene units of said group are optionally and independently replaced. In some embodiments, L is a divalent optionally substituted straight or branched chain C having 1 to 20 heteroatoms1-100A heteroaliphatic group, wherein one or more methylene units of the group are optionally and independently replaced.
In some embodiments, the linker moiety (e.g., L) is or includes one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) polyethylene glycol units. In some embodiments, the linker moiety is or comprises- (CR)2CR2O)nWherein each of R and n is independently as described in the disclosure. In some embodiments, the linker moiety is or comprises- (CH)2CH2O)n-, wherein n is as described in the present disclosure. In some embodiments, one or more methylene units of L are independently replaced by- (CH)2CH2O)n-a permutation. In some embodiments, two or more methylene units of L are independently- (CR)2CR2O)n-or- (CH)2CH2O)n-a permutation. In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, n is 5. In some embodiments, n is 6. In some embodiments, n is 7. In some embodiments, n is 8. In some embodiments, n is 9. In some embodiments, n is 10. In some embodiments, n is 11. In some embodiments, n is 12. In some embodiments, n is 13. In some embodiments, n is 14. In some embodiments, n is 15. In some embodiments, n is 16. In some embodiments, n is 17. In some embodiments, n is 18. In some embodiments, n is 19. In some embodiments, n is 20.
In some embodiments, - (CR) in the linker moiety, e.g., L2CR2Number of O) -cells or-(CH2CH2The number of O) -units is at least about 1 to 20, 2 to 20, 3 to 30, 4 to 20, 5 to 20, 6 to 20, 7 to 20, 8 to 20, 9 to 20, 10 to 20, 11 to 20, 12 to 20, 13 to 20, 14 to 20, 15 to 20 or about or at least about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20. In some embodiments, it is about or at least about 1. In some embodiments, it is about or at least about 2. In some embodiments, it is about or at least about 3. In some embodiments, it is about or at least about 4. In some embodiments, it is about or at least about 5. In some embodiments, it is about or at least about 6. In some embodiments, it is about or at least about 7. In some embodiments, it is about or at least about 8. In some embodiments, it is about or at least about 9. In some embodiments, it is about or at least about 10. In some embodiments, it is about or at least about 11. In some embodiments, it is about or at least about 12. In some embodiments, it is about or at least about 13. In some embodiments, it is about or at least about 14. In some embodiments, it is about or at least about 15. In some embodiments, it is about or at least about 16. In some embodiments, it is about or at least about 17. In some embodiments, it is about or at least about 18. In some embodiments, it is about or at least about 19. In some embodiments, it is about or at least about 20.
In some embodiments, a linker moiety (e.g., L) comprises one or more — (CR) as described herein2CR2O)n-and/or- (CH)2CH2O)n-and one or more amino acid residues.
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001231
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001232
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001233
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001234
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001235
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001236
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001237
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001238
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001241
In some embodiments, the connector sub-portion or L is or includes
Figure BDA0003526174380001242
In some embodiments, the linker moiety (e.g., L) is or includes one or more (e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20 or more) amino acid residues. As used in this disclosure, "one or more"May be 1 to 100, 1 to 50, 1 to 40, 1 to 30, 1 to 20, 1 to 10, 1 to 5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 or more. In some embodiments, one or more methylene units of L are independently replaced with an amino acid residue. In some embodiments, one or more methylene units of L are independently replaced with an amino acid residue, wherein the amino acid residue is an amino acid of formula a-I or a salt thereof. In some embodiments, one or more methylene units of L are independently replaced with an amino acid residue, wherein each amino acid residue independently has-N (R) a1)-La1-C(Ra2)(Ra3)-La2-CO-or a salt form thereof. In some embodiments, the amino acid is a natural amino acid. In some embodiments, the amino acid is glycine. In some embodiments, the amino acid is an unnatural amino acid. In some embodiments, the amino acid is a D-amino acid. In some embodiments, the amino acid is beta-alanine. In some embodiments, the amino acid residue has a structure of-C (O) - (CH)2CH2O)n-CH2CH2NR '-or a salt form thereof, wherein n is 0 to 20 (e.g., 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20), and R' is as described herein. In some embodiments, n is 0. In some embodiments, n is 0 to 12. In some embodiments, n is 1 to 12. In some embodiments, R' is — H.
In some embodiments, linker moieties include one or more moieties, such as amino, carbonyl, and the like, that are available for linking to other moieties. In some embodiments, the linker moiety comprises one or more-NR '-, where R' is as described in the disclosure. In some embodiments, -NR' -improves solubility. In some embodiments, -NR' -acts as a point of attachment to another moiety. In some embodiments, R' is — H. In some embodiments, one or more methylene units of L are independently replaced with-NR '-wherein R' is as described in the disclosure.
In some embodiments, the linker moiety (e.g., L) comprises a-c (o) -group available for attachment to the moiety. In some embodiments, one or more methylene units of L are independently replaced with-c (o) -.
In some embodiments, the linker moiety (e.g., L) comprises an-NR' -group that is available for attachment to the moiety. In some embodiments, one or more methylene units of L are independently replaced by-N (R') -o.
In some embodiments, the linker moiety (e.g., L) comprises a-c (o) NR' -group that is available for attachment to the moiety. In some embodiments, one or more methylene units of L are independently replaced by-c (o) N (R') -L.
In some embodiments, the linker moiety (e.g., L) comprises-C (R')2-a group. In some embodiments, one or more methylene units of L are independently replaced by-C (R')2-a permutation. In some embodiments, -C (R')2-is-CHR' -. In some embodiments, R' is- (CH)2)2C(O)NH(CH2)11COOH. In some embodiments, R' is- (CH)2)2COOH. In some embodiments, R' is — COOH.
In some embodiments, the linker moiety is or includes one or more cyclic moieties, e.g., one or more methylene units of L are replaced by-Cy-. In some embodiments, the linker moiety (e.g., L) comprises an aryl ring. In some embodiments, the linker moiety (e.g., L) comprises a heteroaryl ring. In some embodiments, the linker moiety (e.g., L) comprises an aliphatic ring. In some embodiments, the linker moiety (e.g., L) comprises a heterocyclyl ring. In some embodiments, the linker moiety (e.g., L) comprises multiple rings. In some embodiments, the loops in the linker moiety (e.g., L) are 3 to 20-membered. In some embodiments, the ring is 5-membered. In some embodiments, the ring is 6-membered. In some embodiments, the ring in the linker is the product of a cycloaddition reaction (e.g., click chemistry and variations thereof) used to link different moieties together.
In some embodiments, the connector sub-portion (e.g., L) is or includes
Figure BDA0003526174380001251
In some embodiments, the methylene unit of L is substituted with one or more substituents selected from the group consisting of alkyl, aryl, cycloalkyl, and heteroaryl
Figure BDA0003526174380001252
And (4) replacement. In some embodiments, -Cy-is
Figure BDA0003526174380001253
In some embodiments, the linker moiety (e.g., L) is or comprises-Cy-. In some embodiments, the methylene unit of L is replaced with-Cy-. In some embodiments, -Cy-is
Figure BDA0003526174380001254
In some embodiments, -Cy-is
Figure BDA0003526174380001255
In some embodiments, -Cy-is
Figure BDA0003526174380001256
In some embodiments, the linker moiety (e.g., L) in a provided agent (e.g., a compound in table 1) comprises
Figure BDA0003526174380001261
In some embodiments of the present invention, the,
Figure BDA0003526174380001262
in a structure of
Figure BDA0003526174380001263
Figure BDA0003526174380001264
In some embodiments of the present invention, the,
Figure BDA0003526174380001265
is composed of
Figure BDA0003526174380001266
In some embodiments of the present invention, the,
Figure BDA0003526174380001267
is composed of
Figure BDA0003526174380001268
In some embodiments, the connector sub-portions are as described in table 1. Additional connector sub-portions, for example, including for L2The described sub-portion of the connector. In some embodiments, L is L of the present disclosure1. In some embodiments, L is L as described in this disclosure2. In some embodiments, L is L as described in this disclosure3. In some embodiments, L is L as described in this disclosureb
In some embodiments, L is
Figure BDA0003526174380001269
Figure BDA00035261743800012610
In some embodiments, a linker comprises an amino acid sequence comprising one or more amino acid residues. In some embodiments, the linker is or comprises
Figure BDA00035261743800012611
In some embodiments, the linker is or comprises
Figure BDA0003526174380001271
In some embodiments, the linker is or includes a Gly residue. In some embodiments, the linker is or comprises- (Gly) n-, where n is as described herein. In some embodiments, n is 1 to 10, e.g., 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10. In some embodiments, the linker is or comprises-Gly-. In some embodiments, the linker is or includes-Gly-. Without wishing to be bound by theory, in some embodiments, linkers comprising amino acid residues may provide various moieties that may facilitate, enhance, and/or enhance one or more properties and/or activitiesFractional hardness and/or orientation.
In some embodiments, the linker is linked to a moiety, such as a target binding moiety or an antibody binding moiety, via the N-terminal amino acid residue (e.g., via an amino group) or the C-terminal amino acid residue (e.g., via a-COOH group). In some embodiments, the linker is attached to the moiety via a side chain.
In some embodiments, the linker (e.g., L) is or includes divalent optionally substituted C1-20An aliphatic group. In some embodiments, the linker (e.g., L) is or includes divalent optionally substituted C 1-20An alkylene group. In some embodiments, the divalent group is linear. In some embodiments, the linker (e.g., L) is or comprises a linear- (CH)2)n-, where n is 1 to 20 (e.g. 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20). In some embodiments, the linker (e.g., L) is or includes a linker having NHR' - (CH)2)n-a residue of an amino acid of the structure of-COOH or a salt thereof. In some embodiments, the linker (e.g., L) is or includes-NR' - (CH)2)n-CO-or a salt form thereof. In some embodiments, R' is — H. In some embodiments, as shown herein, the linker comprises an albumin binding moiety.
In some embodiments, as used herein (e.g., in various moieties), n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, n is 5. In some embodiments, n is 6. In some embodiments, n is 7. In some embodiments, n is 8. In some embodiments, n is 9. In some embodiments, n is 10. In some embodiments, n is 11. In some embodiments, n is 12. In some embodiments, n is 13. In some embodiments, n is 14. In some embodiments, n is 15. In some embodiments, n is 16. In some embodiments, n is 17. In some embodiments, n is 18. In some embodiments, n is 19. In some embodiments, n is 20.
In some embodiments, the linker is or includes a moiety, or fragment thereof, between two cyclic peptide moieties of a provided compound, e.g., in table 1.
Some embodiments of variables
For example, exemplary embodiments of variables are described throughout this disclosure. Embodiments of different variables may optionally be combined, as appreciated by those skilled in the art.
ABT is an antibody binding moiety as described herein, as defined above and described herein. In some embodiments, the ABT is an ABT of a compound selected from those depicted in table 1 below. In some embodiments, the ABT is a moiety selected from table a-1. In some embodiments, ABT is a moiety described in table 1.
In some embodiments, L is a bivalent or multivalent linker moiety linking one or more antibody binding moieties to one or more target binding moieties. In some embodiments, L is a bivalent linker moiety linking ABT and TBT. In some embodiments, L is a multivalent linker moiety linking ABT and TBT.
In some embodiments, L is a linker moiety of a compound selected from those depicted in table 1 below.
As defined above and described herein, TBT is a target binding moiety as described herein.
In some embodiments, the TBT is a target binding moiety of a compound selected from those depicted in table 1 below. In some embodiments, the TBT is a moiety selected from Table T-1. In some embodiments, the TBT is part described in table 1.
As defined above and described herein, R1、R3And R5Each of which is independently hydrogen or an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or from 1 to 5 independentlyAn 8-to 10-membered bicyclic heteroaromatic ring selected from a heteroatom of nitrogen, oxygen or sulfur; or: r1And R1' optionally together with its intervening carbon atoms form a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring or a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; r3And R3' optionally together with its intervening carbon atoms form a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring or a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; r bound to the same carbon atom 5Group and R5' the group optionally forms, together with its intervening carbon atoms, a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring or a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or two R5The radicals optionally forming C together with their intermediate atoms1-10A divalent linear or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 3 methylene units of said chain are independently and optionally substituted by-S-, -SS-, -N (R) -, -O-, -C (O) -, -OC (O) -, -C (O) O-, -C (O) N (R) -, -N (R) C (O) -, -S (O)2-or-Cy1-substitution, wherein each-Cy1-independently is a 5-to 6-membered heteroarylene having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R1Is hydrogen. In some embodiments, R1Is an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R 1Is optionally substituted C1-6An aliphatic group. In some embodiments, R1Is an optionally substituted 3-to 8-membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, R1Is optionally substituted phenyl. In some embodiments, R1Is an optionally substituted 8-to 10-membered bicyclic aromatic carbocyclic ring. In some embodiments, R1Is an optionally substituted 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R1Is an optionally substituted 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R1Is an optionally substituted 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R1Is composed of
Figure BDA0003526174380001291
In some embodiments, R1Is composed of
Figure BDA0003526174380001292
In some embodiments, R1Is composed of
Figure BDA0003526174380001293
In some embodiments, R1Is composed of
Figure BDA0003526174380001294
In some embodiments, R1Is composed of
Figure BDA0003526174380001295
In some embodiments, R1Is composed of
Figure BDA0003526174380001296
In some embodiments, R1Is composed of
Figure BDA0003526174380001297
In some embodiments, R1Is composed of
Figure BDA0003526174380001298
In some embodiments, R1Is composed of
Figure BDA0003526174380001299
In some embodiments, R1Is composed of
Figure BDA00035261743800012910
In some embodiments, R 1Is composed of
Figure BDA00035261743800012911
In some embodiments, R1Is composed of
Figure BDA00035261743800012912
In some embodiments, R1Is composed of
Figure BDA00035261743800012913
In some embodiments, R1Is composed of
Figure BDA00035261743800012914
In some embodiments, R1Is composed of
Figure BDA00035261743800012915
In some embodiments, R1Is composed of
Figure BDA0003526174380001301
In some embodiments, R1Is composed of
Figure BDA0003526174380001302
In some embodiments, R1Is composed of
Figure BDA0003526174380001303
In some embodiments, R1Is composed of
Figure BDA0003526174380001304
In some embodiments, R1Is composed of
Figure BDA0003526174380001305
In some embodiments, R1And R1' optionally together with their intervening carbon atoms form a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring. In some embodiments, R1And R1' optionally together with their intervening carbon atoms form a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R1Selected from those depicted in table 1 below.
In some embodiments, R is R as described in the disclosure1. In some embodiments, Ra2Is R as described in this disclosure1. In some embodiments, Ra3Is R as described in this disclosure1
In some embodiments, R3Is hydrogen. In some embodiments, R3Is an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R 3Is optionally substituted C1-6An aliphatic group. In some embodiments, R3Is an optionally substituted 3-to 8-membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, R3Is optionally substituted phenyl. In some embodiments, R3Is an optionally substituted 8-to 10-membered bicyclic aromatic carbocyclic ring. In some embodiments, R3Is an optionally substituted 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R3Is an optionally substituted heteroatom having 1 to 4 heteroatoms independently selected from nitrogen, oxygen or sulfurA 5-to 6-membered monocyclic heteroaromatic ring. In some embodiments, R3Is an optionally substituted 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R3Is methyl. In some embodiments, R3Is composed of
Figure BDA0003526174380001306
In some embodiments, R3Is composed of
Figure BDA0003526174380001307
In some embodiments, R3Is composed of
Figure BDA0003526174380001308
In some embodiments, R3Is composed of
Figure BDA0003526174380001309
In some embodiments, R3Is composed of
Figure BDA0003526174380001311
Wherein the attachment site has (S) stereochemistry. In some embodiments, R3Is composed of
Figure BDA0003526174380001312
Wherein the attachment site has (R) stereochemistry. In some embodiments, R 3Is composed of
Figure BDA0003526174380001313
Wherein the attachment site has (S) stereochemistry. In some embodiments, R3Is composed of
Figure BDA0003526174380001314
Wherein the attachment site has (R) stereochemistry.
In some embodiments, R3Is composed of
Figure BDA0003526174380001315
Wherein the attachment site has (S) stereochemistry. In some embodiments, R3Is composed of
Figure BDA0003526174380001316
Wherein the attachment site has (R) stereochemistry.
In some embodiments, R3And R3' optionally together with their intervening carbon atoms form a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring. In some embodiments, R3And R3' optionally together with their intervening carbon atoms form a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R3Selected from those depicted in table 1 below.
In some embodiments, R is R as described in the disclosure2. In some embodiments, Ra2Is R as described in this disclosure2. In some embodiments, Ra3Is R as described in this disclosure2
In some embodiments, R5Is hydrogen. In some embodiments, R5Is an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R 5Is optionally substituted C1-6An aliphatic group. In some embodiments, R5Is an optionally substituted 3-to 8-membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, R5Is optionally substituted phenyl. In some embodiments, R5Is an optionally substituted 8-to 10-membered bicyclic aromatic carbocyclic ring. In some embodiments, R5Is a compound having 1 to 2 independently selected nitrogen atomsOptionally substituted 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle of a heteroatom of oxygen or sulfur. In some embodiments, R5Is an optionally substituted 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R5Is an optionally substituted 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R5Is methyl. In some embodiments, R5Is composed of
Figure BDA0003526174380001317
In some embodiments, R5Is composed of
Figure BDA0003526174380001321
In some embodiments, R5Is composed of
Figure BDA0003526174380001322
In some embodiments, R5Is composed of
Figure BDA0003526174380001323
In some embodiments, R5Is composed of
Figure BDA0003526174380001324
In some embodiments, R5Is composed of
Figure BDA0003526174380001325
In some embodiments, R5Is composed of
Figure BDA0003526174380001326
In some embodiments, R5Is composed of
Figure BDA0003526174380001327
In some embodiments, R5Is composed of
Figure BDA0003526174380001328
Wherein the connecting position Dots have (S) stereochemistry. In some embodiments, R5Is composed of
Figure BDA0003526174380001329
Wherein the attachment site has (R) stereochemistry. In some embodiments, R5Is composed of
Figure BDA00035261743800013210
Wherein the attachment site has (S) stereochemistry. In some embodiments, R5Is composed of
Figure BDA00035261743800013211
Wherein the attachment site has (R) stereochemistry. In some embodiments, R5Is composed of
Figure BDA00035261743800013212
In some embodiments, R5Is composed of
Figure BDA00035261743800013213
In some embodiments, R5Is composed of
Figure BDA00035261743800013214
In some embodiments, R5Is composed of
Figure BDA00035261743800013215
In some embodiments, R5Is composed of
Figure BDA00035261743800013216
In some embodiments, R5Is composed of
Figure BDA00035261743800013217
In some embodiments, R5Is composed of
Figure BDA00035261743800013218
In some embodiments, R5Is composed of
Figure BDA00035261743800013219
In some embodiments, R5Is composed of
Figure BDA00035261743800013220
In some embodiments, R5Is composed of
Figure BDA00035261743800013221
In some embodiments, R5Is composed of
Figure BDA00035261743800013222
In some embodiments, R5Is composed of
Figure BDA00035261743800013223
In some embodiments, R5Is composed of
Figure BDA00035261743800013224
In some embodiments, R5Is composed of
Figure BDA00035261743800013225
In some embodiments, R5Is composed of
Figure BDA0003526174380001331
In some embodiments, R5Is composed of
Figure BDA0003526174380001332
In some embodiments, R5Is composed of
Figure BDA0003526174380001333
In some embodiments, R5Is composed of
Figure BDA0003526174380001334
In some embodiments, R5Is composed of
Figure BDA0003526174380001335
In some embodiments, R5Is composed of
Figure BDA0003526174380001336
In some embodiments, R5Is composed of
Figure BDA0003526174380001337
In some embodiments, R4Is 5
Figure BDA0003526174380001338
In some embodiments, R5Is composed of
Figure BDA0003526174380001339
In some embodiments, R5Is composed of
Figure BDA00035261743800013310
In some embodiments, R5Is composed of
Figure BDA00035261743800013311
In some embodiments, R5Is composed of
Figure BDA00035261743800013312
In some embodiments, R 4Is composed of
Figure BDA00035261743800013313
Wherein the attachment site has (S) stereochemistry. In some embodiments, R4Is composed of
Figure BDA00035261743800013314
Wherein the attachment site has (R) stereochemistry.
In some embodiments, R is attached to the same carbon atom5And R5The' group optionally forms a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring together with its intervening carbon atoms. In some embodiments, R is attached to the same carbon atom5And R5The' group optionally together with its intervening carbon atoms forms a compound having 1 to 2 substituents independently selected from nitrogenA 4-to 8-membered saturated or partially unsaturated spiroheterocyclic ring of a heteroatom of oxygen or sulfur.
In some embodiments, two R5The radicals together with their central atoms forming C1-10A divalent linear or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 3 methylene units of said chain are independently and optionally substituted by-S-, -SS-, -N (R) -, -O-, -C (O) -, -OC (O) -, -C (O) O-, -C (O) N (R) -, -N (R) C (O) -, -S (O)2-or-Cy1-substitution, wherein each-Cy1-independently is a 5-to 6-membered heteroarylene having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, two R5The radicals together with their central atoms forming
Figure BDA00035261743800013315
In some embodiments, two R5The radicals together with their central atoms forming
Figure BDA00035261743800013316
In some embodiments, two R5The radicals together with their central atoms forming
Figure BDA00035261743800013317
In some embodiments, two R5The radicals together with their central atoms forming
Figure BDA0003526174380001341
In some embodiments, R5Selected from those depicted in table 1 below.
In some embodiments, R is R as described in the disclosure5. In some embodiments, Ra2Is R as described in this disclosure5. In some embodiments, Ra3Is R as described in this disclosure5
As defined above and described herein, R1′、R3' and R5Each of' is independently hydrogen or C1-3An aliphatic group.
In some embodiments, R1' is hydrogen. In some embodiments, R1' is C1-3An aliphatic group.
In some embodiments, R1' is methyl. In some embodiments, R1' is ethyl. In some embodiments, R1' is n-propyl. In some embodiments, R1' is isopropyl. In some embodiments, R1' is cyclopropyl.
In some embodiments, R1' is selected from those depicted in table 1 below.
In some embodiments, R3' is hydrogen. In some embodiments, R3' is C1-3An aliphatic group.
In some embodiments, R3' is methyl. In some embodiments, R3' is ethyl. In some embodiments, R 3' is n-propyl. In some embodiments, R3' is isopropyl. In some embodiments, R3' is cyclopropyl.
In some embodiments, R3' is selected from those depicted in table 1 below.
In some embodiments, R5' is hydrogen. In some embodiments, R5' is C1-3An aliphatic group.
In some embodiments, R5' is methyl. In some embodiments, R5' is ethyl. In some embodiments, R5' is n-propyl. In some embodiments, R5' is isopropyl. In some embodiments, R5' is cyclopropyl.
In some embodiments, R5' is selected from those depicted in table 1 below.
As defined above and described herein, R2、R4And R6Each of which is independently hydrogen or C1-4An aliphatic group, or: r2And R1Optionally together with their intermediate atoms form a 4-to 8-membered saturated ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfurOr a partially unsaturated monocyclic heterocycle; r4And R3Optionally together with their intermediate atoms, form a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or R6Group and R adjacent thereto5The groups optionally form, together with their intermediate atoms, a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R2Is hydrogen. In some embodiments, R2Is C1-4An aliphatic group. In some embodiments, R2Is methyl. In some embodiments, R2Is ethyl. In some embodiments, R2Is n-propyl. In some embodiments, R2Is isopropyl. In some embodiments, R2Is n-butyl. In some embodiments, R2Is an isobutyl group. In some embodiments, R2Is a tert-butyl group.
In some embodiments, R2And R1Together with their intermediate atoms form a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R2And R1Together with their central atoms form
Figure BDA0003526174380001351
In some embodiments, R2And R1Together with their central atoms form
Figure BDA0003526174380001352
In some embodiments, R2Selected from those depicted in table 1 below.
In some embodiments, R4Is hydrogen. In some embodiments, R4Is C1-4An aliphatic group. In some embodiments, R4Is methyl. In some embodiments, R4Is ethyl. In some embodiments, R4Is n-propyl. In some embodiments, R4Is isopropyl. In some embodiments, R4Is n-butyl. In some embodiments, R 4Is an isobutyl group. In some embodiments, R4Is a tert-butyl group.
In some embodiments, R4And R3Together with their intermediate atoms form a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R4And R3Together with their central atoms form
Figure BDA0003526174380001353
In some embodiments, R4And R3Together with their central atoms form
Figure BDA0003526174380001354
In some embodiments, R4Selected from those depicted in table 1 below.
In some embodiments, R6Is hydrogen. In some embodiments, R6Is C1-4An aliphatic group. In some embodiments, R6Is methyl. In some embodiments, R6Is ethyl. In some embodiments, R6Is n-propyl. In some embodiments, R6Is isopropyl. In some embodiments, R6Is n-butyl. In some embodiments, R6Is an isobutyl group. In some embodiments, R6Is a tert-butyl group.
In some embodiments, R6Group and R adjacent thereto5The groups together with their intervening atoms form a 4-to 8-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R6Group and R adjacent thereto5The radicals together with their central atoms forming
Figure BDA0003526174380001355
In some embodiments, R6Group and R adjacent thereto5The radicals together with their central atoms forming
Figure BDA0003526174380001356
In some embodiments, R6Selected from those depicted in table 1 below.
In some embodiments, R is R as described in the disclosure1'. In some embodiments, Ra2Is R as described in this disclosure1'. In some embodiments, Ra3Is R as described in this disclosure1'. In some embodiments, R is R as described in the disclosure3'. In some embodiments, Ra2Is R as described in this disclosure3'. In some embodiments, Ra3Is R as described in this disclosure3'. In some embodiments, R is R as described in the disclosure2. In some embodiments, Ra2Is R as described in this disclosure2. In some embodiments, Ra3Is R as described in this disclosure2. In some embodiments, R is R as described in the disclosure4. In some embodiments, Ra2Is R as described in this disclosure4. In some embodiments, Ra3Is R as described in this disclosure4. In some embodiments, R is R as described in the disclosure6. In some embodiments, Ra2Is R as described in this disclosure6. In some embodiments, R a3Is R as described in this disclosure6
As defined above and described herein, L1To connect to
Figure BDA0003526174380001361
Figure BDA0003526174380001362
A trivalent linker moiety of (a).
In some embodiments, L1Is composed of
Figure BDA0003526174380001363
In some embodiments, L1Is composed of
Figure BDA0003526174380001364
In some embodiments, L1Is composed of
Figure BDA0003526174380001365
In some embodiments, L1Is composed of
Figure BDA0003526174380001371
In some embodiments, L1Is composed of
Figure BDA0003526174380001372
In some embodiments, L1Is composed of
Figure BDA0003526174380001373
In some embodiments, L1Is composed of
Figure BDA0003526174380001374
In some embodiments, L1Is composed of
Figure BDA0003526174380001375
In some embodiments, L1Is composed of
Figure BDA0003526174380001381
In some embodiments, L1Is composed of
Figure BDA0003526174380001382
In some embodiments, L1Is composed of
Figure BDA0003526174380001383
In some embodiments, L1Is composed of
Figure BDA0003526174380001384
In some embodimentsIn, L1Is composed of
Figure BDA0003526174380001385
In some embodiments, L1Is composed of
Figure BDA0003526174380001391
In some embodiments, L1Is composed of
Figure BDA0003526174380001392
In some embodiments, L1Is composed of
Figure BDA0003526174380001393
In some embodiments, L1Is composed of
Figure BDA0003526174380001401
In some embodiments, L1Is composed of
Figure BDA0003526174380001402
In some embodiments, L1Is composed of
Figure BDA0003526174380001403
In some embodiments, L1Is composed of
Figure BDA0003526174380001411
In some embodiments, L1Is composed of
Figure BDA0003526174380001412
In some embodiments, L1Is composed of
Figure BDA0003526174380001413
In some embodiments, L1Selected from those depicted in table 1 below.
As defined above and described herein, L2Is a covalent bond or C1-10A divalent linear or branched saturated or unsaturated hydrocarbon chain, wherein 1 of said chainUp to 3 methylene units are independently and optionally substituted by-S-, -N (R) -, -O-, -C (O) -, -OC (O) -, -C (O) O-, -C (O) N (R) -, -N (R) C (O) -, -S (O) 2-、
Figure BDA0003526174380001414
Figure BDA0003526174380001415
or-Cy1-substitution, wherein each-Cy1-independently is a 5-to 6-membered heteroarylene having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, L2Is a covalent bond. In some embodiments, L2Is C1-10A divalent linear or branched saturated or unsaturated hydrocarbon chain, wherein 1 to 3 methylene units of said chain are independently and optionally substituted by-S-, -N (R) -, -O-, -C (O) -, -OC (O) -, -C (O) O-, -C (O) N (R) -, -N (R) C (O) -, -S (O)2-、
Figure BDA0003526174380001416
Figure BDA0003526174380001417
or-Cy1-substitution, wherein each-Cy1-independently is a 5-to 6-membered heteroarylene having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, L2Is composed of
Figure BDA0003526174380001421
In some embodiments, L2Is composed of
Figure BDA0003526174380001422
In some embodiments, L2Is composed of
Figure BDA0003526174380001423
In some embodiments, L2Is composed of
Figure BDA0003526174380001424
In some embodiments, L2Is composed of
Figure BDA0003526174380001425
In some embodiments, L2Is composed of
Figure BDA0003526174380001426
In some embodiments, L2Selected from those depicted in table 1 below.
In some embodiments, L is L as described in this disclosure2
TBT is a target binding moiety as defined above and described herein.
In some embodiments, the TBT is a target binding moiety.
In some embodiments, the TBT is
Figure BDA0003526174380001427
In some embodiments, the TBT is
Figure BDA0003526174380001428
In some embodiments, the TBT is selected from those depicted in table 1 below.
As defined above and described herein, each of m and n is independently 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10.
In some embodiments, m is 1. In some embodiments, m is 2. In some embodiments, m is 3. In some embodiments, m is 4. In some embodiments, m is 5. In some embodiments, m is 6. In some embodiments, m is 7. In some embodiments, m is 8. In some embodiments, m is 9. In some embodiments, m is 10.
In some embodiments, m is selected from those depicted in table 1 below.
In some embodiments, n is 1. In some embodiments, n is 2. In some embodiments, n is 3. In some embodiments, n is 4. In some embodiments, n is 5. In some embodiments, n is 6. In some embodiments, n is 7. In some embodiments, n is 8. In some embodiments, n is 9. In some embodiments, n is 10.
In some embodiments, n is selected from those depicted in table 1 below.
As defined above and described herein, R7Each of which is independently hydrogen or an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or: r bound to the same carbon atom 7Group and R7The' group optionally forms, together with its intervening carbon atoms, a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring or a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R7Is hydrogen. In some embodiments, R7Is an optionally substituted group selected from: c1-6An aliphatic group; a 3 to 8 membered saturated or partially unsaturated monocyclic carbocyclic ring; a phenyl group; an 8 to 10 membered bicyclic aromatic carbocyclic ring; a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur; a 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; or an 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R7Is optionally substituted C1-6An aliphatic group. In some embodiments, R7Is an optionally substituted 3-to 8-membered saturated or partially unsaturated monocyclic carbocyclic ring. In some embodiments, R7Is optionally substituted phenyl. In some embodiments, R7Is an optionally substituted 8-to 10-membered bicyclic aromatic carbocyclic ring. In some embodiments, R 7Is an optionally substituted 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R7Is an optionally substituted 5-to 6-membered monocyclic heteroaromatic ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R7Is an optionally substituted 8-to 10-membered bicyclic heteroaromatic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R7Is methyl. In some embodiments, R7Is composed of
Figure BDA0003526174380001431
In some embodiments, R7Is composed of
Figure BDA0003526174380001432
In some embodiments, R7Is composed of
Figure BDA0003526174380001433
In some embodiments, R7Is composed of
Figure BDA0003526174380001434
In some embodiments, R7Is composed of
Figure BDA0003526174380001435
In some embodiments, R7Is composed of
Figure BDA0003526174380001436
In some embodiments, R7Is composed of
Figure BDA0003526174380001441
In some embodiments, R7Is composed of
Figure BDA0003526174380001442
In some embodiments, R7Is composed of
Figure BDA0003526174380001443
In some embodiments, R7Is composed of
Figure BDA0003526174380001444
In some embodiments, R7Is composed of
Figure BDA0003526174380001445
In some embodiments, R7Is composed of
Figure BDA0003526174380001446
In some embodiments, R7Is composed of
Figure BDA0003526174380001447
In some embodiments, R7Is composed of
Figure BDA0003526174380001448
In some embodiments, R7Is composed of
Figure BDA0003526174380001449
In some embodiments, R7Is composed of
Figure BDA00035261743800014410
In some embodiments, R7Is composed of
Figure BDA00035261743800014411
In some embodiments, R7Is composed of
Figure BDA00035261743800014412
In some embodiments, R7Is composed of
Figure BDA00035261743800014413
In some embodiments, R 7Is composed of
Figure BDA00035261743800014414
In some embodiments, R is attached to the same carbon atom7Group and R7' the groups together with their intervening carbon atoms form a 3-to 8-membered saturated or partially unsaturated spirocyclic carbocyclic ring. In some embodiments, R is attached to the same carbon atom7Group and R7The' group together with its intervening carbon atoms forms a 4-to 8-membered saturated or partially unsaturated spirocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R7Selected from those depicted in table 1 below.
As defined above and described herein, R7Each of' is independently hydrogen or C1-3An aliphatic group.
In some embodiments, R7' is hydrogen. In some embodiments, R7' is methyl. In some embodiments, R7' is ethyl. In some embodiments, R7' is n-propyl. In some embodiments, R7' is isopropyl.
In some embodiments, R7' is selected from those depicted in table 1 below.
As defined above and described herein, R8Each of which is independently hydrogen or C1-4An aliphatic group, or: r8Group and R adjacent thereto7The groups optionally form, together with their intermediate atoms, a 4-to 8-membered saturated or partially unsaturated monocyclic heterocycle having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R8Is hydrogen. In some embodiments, R8Is C1-4An aliphatic group. In some embodiments, R8Is methyl. In some embodiments, R8Is ethyl. In some embodiments, R8Is n-propyl. In some embodiments, R8Is isopropyl. In some embodiments, R8Is n-butyl. In some embodiments, R8Is an isobutyl group. In some embodiments, R8Is a tert-butyl group.
In some embodiments, R8Group and R adjacent thereto7The groups together with their intervening atoms form a 4-to 8-membered saturated or partially unsaturated monocyclic heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
In some embodiments, R8Group and R adjacent thereto7The radicals together with their central atoms forming
Figure BDA0003526174380001451
In some embodiments, R8Group and R adjacent thereto7The radicals together with their central atoms forming
Figure BDA0003526174380001452
In some embodiments, R8Selected from those depicted in table 1 below.
As defined above and described herein, R9Is hydrogen, C1-3Aliphatic radical or-C (O) C1-3An aliphatic group.
In some embodiments, R9Is hydrogen. In some embodiments, R9Is C1-3An aliphatic group. In some embodiments, R9is-C (O) C1-3An aliphatic group.
In some embodiments, R 9Is methyl. In some embodiments, R9Is ethyl. In some embodiments, R9Is n-propyl. In some embodiments, R9Is isopropyl. In some embodiments, R9Is cyclopropyl. In some embodiments, R9is-C (O) Me. In some embodiments, R9is-C (O) Et. In some embodiments, R9is-C (O) CH2CH2CH3. In some embodiments, R9is-C (O) CH (CH)3)2. In some embodiments, R9is-C (O) cyclopropyl.
In some embodiments, R9Selected from those depicted in table 1 below.
In some embodiments, R is R as described in the disclosure7. In some embodiments,Ra2Is R as described in this disclosure7. In some embodiments, Ra3Is R as described in this disclosure7. In some embodiments, R is R as described in the disclosure7'. In some embodiments, Ra2Is R as described in this disclosure7'. In some embodiments, Ra3Is R as described in this disclosure7'. In some embodiments, R is R as described in the disclosure8. In some embodiments, Ra2Is R as described in this disclosure8. In some embodiments, Ra3Is R as described in this disclosure8. In some embodiments, R is R as described in the disclosure 8'. In some embodiments, Ra2Is R as described in this disclosure8'. In some embodiments, Ra3Is R as described in this disclosure8'. In some embodiments, R is R as described in the disclosure9. In some embodiments, Ra2Is R as described in this disclosure9. In some embodiments, Ra3Is R as described in this disclosure9
As defined above and described herein, L3To connect to
Figure BDA0003526174380001461
A divalent linker moiety to TBT.
In some embodiments, L3To connect to
Figure BDA0003526174380001462
A divalent linker moiety to TBT.
In some embodiments, L3Is composed of
Figure BDA0003526174380001463
In some embodiments, L3Is composed of
Figure BDA0003526174380001464
In some embodiments, L3Is composed of
Figure BDA0003526174380001465
In some embodiments, L3Is composed of
Figure BDA0003526174380001466
In some embodiments, L3Is composed of
Figure BDA0003526174380001467
In some embodiments, L3Is composed of
Figure BDA0003526174380001468
In some embodiments, L3Selected from those depicted in table 1 below.
In some embodiments, L is L as described in this disclosure3
O is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 as defined above and described herein.
In some embodiments, o is 1. In some embodiments, o is 2. In some embodiments, o is 3. In some embodiments, o is 4. In some embodiments, o is 5. In some embodiments, o is 6. In some embodiments, o is 7. In some embodiments, o is 8. In some embodiments, o is 9. In some embodiments, o is 10.
In some embodiments, o is selected from those depicted in table 1 below.
In certain embodiments, the present disclosure provides a compound of formula II, wherein L2Is composed of
Figure BDA0003526174380001471
And TBT is
Figure BDA0003526174380001472
Thereby forming a compound of formula II-a:
Figure BDA0003526174380001473
or a pharmaceutically acceptable salt thereof, wherein L1、R1、R1′、R2、R3、R3′、R4、R5、R5′、R6And each of m is as defined above and as described in the examples herein, individually and in combination.
In certain embodiments, the present disclosure provides a compound of formula II, wherein L2Is composed of
Figure BDA0003526174380001474
And TBT is
Figure BDA0003526174380001475
Thereby forming a compound of formula II-b:
Figure BDA0003526174380001481
or a pharmaceutically acceptable salt thereof, wherein L1、R1、R1′、R2、R3、R3′、R4、R5、R5′、R6And each of m is as defined above and as described in the examples herein, individually and in combination.
In certain embodiments, the present disclosure provides a compound of formula II, wherein L2Is composed of
Figure BDA0003526174380001482
And TBT is
Figure BDA0003526174380001483
Thereby forming a compound of formula II-c:
Figure BDA0003526174380001484
or a pharmaceutically acceptable salt thereof, wherein L1、R1、R1′、R2、R3、R3′、R4、R5、R5′、R6And each of m is as defined above and as described in the examples herein, individually and in combination.
In certain embodiments, the present disclosure provides a compound of formula II, wherein L2Is composed of
Figure BDA0003526174380001491
And TBT is
Figure BDA0003526174380001492
Thereby forming a compound of formula II-d:
Figure BDA0003526174380001493
or a pharmaceutically acceptable salt thereof, wherein L1、R1、R1′、R2、R3、R3′、R4、R5、R5′、R6And each of m is as defined above and as described in the examples herein, individually and in combination.
In certain embodiments, the present disclosure provides a compound of formula II, wherein L2Is composed of
Figure BDA0003526174380001494
And TBT is
Figure BDA0003526174380001495
Thereby forming a compound of formula II-e:
Figure BDA0003526174380001496
or a pharmaceutically acceptable salt thereof, wherein L1、R1、R1′、R2、R3、R3′、R4、R5、R5′、R6And each of mOne individually and in combination is as defined above and as described in the examples herein.
In certain embodiments, the present disclosure provides a compound of formula II, wherein L2Is composed of
Figure BDA0003526174380001501
And TBT is
Figure BDA0003526174380001502
Thereby forming a compound of formula II-f:
Figure BDA0003526174380001503
or a pharmaceutically acceptable salt thereof, wherein L1、R1、R1′、R2、R3、R3′、R4、R5、R5′、R6And each of m is as defined above and as described in the examples herein, individually and in combination.
In some embodiments, Ra1Is R as described in this disclosure. In some embodiments, Ra1Is optionally substituted C1-4An aliphatic group. In some embodiments, Ra1Is optionally substituted C1-4An alkyl group. In some embodiments, Ra1Is methyl.
In some embodiments, La1Is L as described in this disclosurea. In some embodiments, La1Is a covalent bond.
In some embodiments, La2Is L as described in this disclosurea. In some embodiments, La2Is a covalent bond.
In some embodiments, LTIs L as described hereina. In some embodiments, LTIs L as described herein. In some embodiments, L TIs a covalent bond. In some embodiments, LTis-CH2-C (O) -. In some embodiments, LTTo the side chain-S- (e.g. via-CH)2) With amino acid residues (e.g. via-C (O)).
In some embodiments, LaIs a covalent bond. In some embodiments, LaIs selected from C1-C10Aliphatic radical or C having 1 to 5 hetero atoms1-C10A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution. In some embodiments, LaIs selected from C1-C5Aliphatic radical or C having 1 to 5 hetero atoms1-C5A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution. In some embodiments, LaIs optionally substituted divalent C1-C5Aliphatic radical, in which one or more methylene units of the radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution. In some embodiments, LaIs an optionally substituted divalent C1-C5 aliphatic group. In some embodiments, LaIs an optionally substituted divalent C having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur 1-C5A heteroaliphatic group.
In some embodiments, Ra2Is R as described in this disclosure. In some embodiments, Ra2Is the side chain of natural amino acid. In some embodiments, Ra3Is R as described in this disclosure. In some embodiments, Ra3Is the side chain of natural amino acid. In some embodiments, R2aAnd R3aOne of which is hydrogen. In some embodiments, Ra2And/or Ra3Is R, wherein R is optionally substituted C1-8Aliphatic or aryl groups. In some embodiments, R is optionally substituted linear C2-8An alkyl group. In some embodiments, R is linear C2-8An alkyl group. In some embodiments, R is an optionally substituted branched chain C2-8An alkyl group. In some embodiments, R is a branched chain C2-8An alkyl group. In some embodiments, R is n-pentyl. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is optionally substituted-CH2-phenyl. In some embodiments, R is 4-phenylphenyl-CH2-。
In some embodiments, each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from: c3-20A cycloaliphatic ring; c6-20An aryl ring; a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 20 membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, each-Cy-is independently an optionally substituted divalent group selected from: c 3-20A cycloaliphatic ring; c6-20An aryl ring; a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 20 membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, -Cy-is as in the present disclosure, e.g., for R and CyLOptionally substituted rings are described, but are divalent.
In some embodiments, -Cy-is monocyclic. In some embodiments, -Cy-is bicyclic. In some embodiments, -Cy-is polycyclic. In some embodiments, -Cy-is saturated. In some embodiments, -Cy-is partially unsaturated. In some embodiments, -Cy-is aromatic. In some embodiments, -Cy-comprises a saturated monocyclic moiety. In some embodiments, -Cy-comprises a partially unsaturated monocyclic moiety. In some embodiments, -Cy-comprises an aromatic monocyclic moiety. In some embodiments, -Cy-comprises a combination of saturated, partially unsaturated, and/or aromatic cyclic moieties. In some embodiments, -Cy-is or comprises a 3-membered ring. In some embodiments, -Cy-is or comprises a 4-membered ring. In some embodiments, -Cy-is or comprises a 5-membered ring. In some embodiments, -Cy-is or comprises a 6-membered ring. In some embodiments, -Cy-is or comprises a 7-membered ring. In some embodiments, -Cy-is or comprises an 8-membered ring. In some embodiments, -Cy-is or includes a 9-membered ring. In some embodiments, -Cy-is or comprises a 10-membered ring. In some embodiments, -Cy-is or comprises an 11-membered ring. In some embodiments, -Cy-is or includes a 12-membered ring. In some embodiments, -Cy-is or comprises a 13-membered ring. In some embodiments, -Cy-is or comprises a 14-membered ring. In some embodiments, -Cy-is or comprises a 15-membered ring. In some embodiments, -Cy-is or comprises a 16-membered ring. In some embodiments, -Cy-is or comprises a 17-membered ring. In some embodiments, -Cy-is or comprises an 18-membered ring. In some embodiments, -Cy-is or comprises a 19-membered ring. In some embodiments, -Cy-is or comprises a 20-membered ring.
In some embodiments, -Cy-is or includes an optionally substituted divalent C3-20A cycloaliphatic ring. In some embodiments, -Cy-is or includes an optionally substituted divalent saturated C3-20A cycloaliphatic ring. In some embodiments, -Cy-is or includes an optionally substituted divalent moiety unsaturated C3-20A cycloaliphatic ring. In some embodiments, -Cy-H is an optionally substituted cycloaliphatic as described in the present disclosure, e.g., a cycloaliphatic example of R.
In some embodiments, -Cy-is or includes optionally substituted C6-20An aryl ring. In some embodiments, -Cy-is or includes optionally substituted phenylene. In some embodiments, -Cy-is or includes optionally substituted 1, 2-phenylene. In some embodiments, -Cy-is or includes optionally substituted 1, 3-phenylene. In some embodiments, -Cy-is or includes being optionally takenSubstituted 1, 4-phenylene. In some embodiments, -Cy-is or includes an optionally substituted divalent naphthalene ring. In some embodiments, -Cy-H is an optionally substituted aryl as described in this disclosure, e.g., an aryl embodiment of R.
In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, and sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heteroaryl ring having 1 to 4 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heteroaryl ring having 1 to 3 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heteroaryl ring having 1 to 2 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heteroaryl ring having one heteroatom independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-H is heteroaryl optionally substituted as described in this disclosure, e.g., heteroaryl embodiments of R. In some embodiments, -Cy-is
Figure BDA0003526174380001521
In some embodiments, -Cy-is or includes an optionally substituted divalent 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, -Cy-is or includes an optionally substituted divalent 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, and sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 3-to 6-membered heterocyclyl ring having 1 to 4 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heterocyclyl ring having 1 to 4 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heterocyclyl ring having 1 to 3 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heterocyclyl ring having 1 to 2 heteroatoms independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted divalent 5-to 6-membered heterocyclyl ring having one heteroatom independently selected from oxygen, nitrogen, sulfur. In some embodiments, -Cy-is or includes an optionally substituted saturated divalent heterocyclic group. In some embodiments, -Cy-is or includes an optionally substituted partially unsaturated divalent heterocyclic group. In some embodiments, -Cy-H is an optionally substituted heterocyclyl as described in this disclosure, e.g., heterocyclyl embodiments of R.
In some embodiments, -Cy-is
Figure BDA0003526174380001531
In some embodiments, -Cy-is
Figure BDA0003526174380001532
In some embodiments, -Cy-is
Figure BDA0003526174380001533
In some embodiments, -Cy-is
Figure BDA0003526174380001534
In some embodiments, -Cy-is
Figure BDA0003526174380001535
In some embodiments, each Xaa is independently an amino acid residue. In some embodiments, each Xaa is independently an amino acid residue of an amino acid of formula a-I.
In some embodiments, t is 0. In some embodiments, t is 1 to 50. In some embodiments, t is z as described in this disclosure.
In some embodiments, y is 1. In some embodiments, y is 2. In some embodiments, y is 3. In some embodiments, y is 4. In some embodiments, y is 5. In some embodiments, y is 6. In some embodiments, y is 7. In some embodiments, y is 8. In some embodiments, y is 9. In some embodiments, y is 10. In some embodiments, y is 11. In some embodiments, y is 12. In some embodiments, y is 13. In some embodiments, y is 14. In some embodiments, y is 15. In some embodiments, y is 16. In some embodiments, y is 17. In some embodiments, y is 18. In some embodiments, y is 19. In some embodiments, y is 20. In some embodiments, y is greater than 20.
In some embodiments, z is 1. In some embodiments, z is 2. In some embodiments, z is 3. In some embodiments, z is 4. In some embodiments, z is 5. In some embodiments, z is 6. In some embodiments, z is 7. In some embodiments, z is 8. In some embodiments, z is 9. In some embodiments, z is 10. In some embodiments, z is 11. In some embodiments, z is 12. In some embodiments, z is 13. In some embodiments, z is 14. In some embodiments, z is 15. In some embodiments, z is 16. In some embodiments, z is 17. In some embodiments, z is 18. In some embodiments, z is 19. In some embodiments, z is 20. In some embodiments, z is greater than 20.
In some embodiments, RcIs R' as described in this disclosure. In some embodiments, RcIs R as described in this disclosure. In some embodiments, Rcis-N (R')2Wherein each R' is independently as described in the disclosure. In some embodiments, Rcis-NH2. In some embodiments, RcIs R-C (O) -, wherein R is as described in the disclosure. In some embodiments, R cis-H.
In some embodiments, a is 1. In some embodiments, a is 2 to 100. In some embodiments, a is 5. In some embodiments, a is 10. In some embodiments, a is 20. In some embodiments, a is 50.
In some embodiments, b is 1. In some embodiments, b is 2 to 100. In some embodiments, b is 5. In some embodiments, b is 10. In some embodiments, b is 20. In some embodiments, b is 50.
In some embodiments, a1 is 0. In some embodiments, a1 is 1.
In some embodiments, a2 is 0. In some embodiments, a2 is 1.
In some embodiments, LbIs L as described in this disclosurea. In some embodiments, Lbincluding-Cy-. In some embodiments, LbIncluding double bonds. In some embodiments, Lbincluding-S-. In some embodiments, Lbincluding-S-S-. In some embodiments, Lbcomprising-C (O) -N (R') -.
In some embodiments, R' is-R, -C (O) OR, OR-S (O)2R, wherein R is as described in the disclosure. In some embodiments, R' is R, wherein R is as described in the disclosure. In some embodiments, R' is-c (o) R, wherein R is as described in the disclosure. In some embodiments, R' is-c (o) OR, wherein R is as described in the disclosure. In some embodiments, R' is-S (O) 2R, wherein R is as described in the disclosure. In some embodiments, R' is hydrogen. In some embodiments, R' is not hydrogen. In some embodiments, R' is R, wherein R is optionally substituted C as described in the disclosure1-20An aliphatic group. In some embodiments, R' is R, wherein R is optionally substituted C as described in the disclosure1-20A heteroaliphatic group. In some embodiments, R' is R, wherein R is optionally substituted C as described in the disclosure6-20And (4) an aryl group. In some embodiments, R' is R, wherein R is optionally substituted C as described in the disclosure6-20An arylaliphatic group. In some embodiments, R' is R, wherein R is optionally substituted C as described in the disclosure6-20An aryl heteroaliphatic group. In some embodiments, R' is R, wherein R is optionally taken as described in the disclosureSubstituted 5 to 20 membered heteroaryl. In some embodiments, R' is R, wherein R is an optionally substituted 3-to 20-membered heterocyclyl as described in this disclosure. In some embodiments, two or more R' are R, and optionally and independently together form an optionally substituted ring as described in the present disclosure.
In some embodiments, each R is independently-H, or an optionally substituted group selected from: c 1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, each R is independently-H, or an optionally substituted group selected from: c 1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; having 1 to 10 substituents independently selected from oxygen, nitrogen5-to 30-membered heteroaryl of heteroatoms of sulfur, phosphorus and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, each R is independently-H, or an optionally substituted group selected from: c 1-20An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-20A heteroaliphatic group; c6-20An aryl group; c6-20An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-20An aryl heteroaliphatic group; a 5-to 20-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 20-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 20-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 20-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, each R is independently-H, or an optionally substituted group selected from: c 1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, each R is independently-H, or an optionally substituted group selected from: c1-20An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-20A heteroaliphatic group; c6-20An aryl group; c6-20An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-20An aryl heteroaliphatic group; a 5-to 20-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 20-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, R is hydrogen. In some embodiments, R is not hydrogen. In some embodiments, R is an optionally substituted group selected from: c 1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; a 5-to 30-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 30 membered heterocyclic ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, R is hydrogen or an optionally substituted group selected from: c1-20An aliphatic group; a phenyl group; a 3 to 7 membered saturated or partially unsaturated carbocyclic ring; 8 to 10 yuanA saturated, partially unsaturated, or aryl ring; a 5-to 6-membered monocyclic heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 4-to 7-membered saturated or partially unsaturated heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur; a 7-to 10-membered bicyclic saturated or partially unsaturated heterocycle having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur; or an 8-to 10-membered bicyclic heteroaryl ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, R is optionally substituted C1-30An aliphatic group. In some embodiments, R is optionally substituted C1-20An aliphatic group. In some embodiments, R is optionally substituted C 1-15An aliphatic group. In some embodiments, R is optionally substituted C1-10An aliphatic group. In some embodiments, R is optionally substituted C1-6An aliphatic group. In some embodiments, R is optionally substituted C1-6An alkyl group. In some embodiments, R is optionally substituted hexyl, pentyl, butyl, propyl, ethyl, or methyl. In some embodiments, R is an optionally substituted hexyl. In some embodiments, R is optionally substituted pentyl. In some embodiments, R is optionally substituted butyl. In some embodiments, R is optionally substituted propyl. In some embodiments, R is optionally substituted ethyl. In some embodiments, R is optionally substituted methyl. In some embodiments, R is hexyl. In some embodiments, R is pentyl. In some embodiments, R is butyl. In some embodiments, R is propyl. In some embodiments, R is ethyl. In some embodiments, R is methyl. In some embodiments, R is isopropyl. In some embodiments, R is n-propyl. In some embodiments, R is tert-butyl. In some embodiments, R is sec-butyl. In some embodiments, R is n-butyl. In some embodiments, R is- (CH) 2)2CN。
In some embodiments, R is optionally substituted C3-30A cycloaliphatic radical. In some embodiments, R is optionally substituted C3-20A cycloaliphatic radical. In some embodiments of the present invention, the,r is optionally substituted C3-10A cycloaliphatic radical. In some embodiments, R is optionally substituted cyclohexyl. In some embodiments, R is cyclohexyl. In some embodiments, R is an optionally substituted cyclopentyl. In some embodiments, R is cyclopentyl. In some embodiments, R is optionally substituted cyclobutyl. In some embodiments, R is cyclobutyl. In some embodiments, R is optionally substituted cyclopropyl. In some embodiments, R is cyclopropyl.
In some embodiments, R is an optionally substituted 3-to 30-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is an optionally substituted 3-to 7-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is an optionally substituted 3-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is an optionally substituted 4-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is an optionally substituted 5-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is an optionally substituted 6-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is an optionally substituted 7-membered saturated or partially unsaturated carbocyclic ring. In some embodiments, R is optionally substituted cycloheptyl. In some embodiments, R is cycloheptyl. In some embodiments, R is optionally substituted cyclohexyl. In some embodiments, R is cyclohexyl. In some embodiments, R is an optionally substituted cyclopentyl. In some embodiments, R is cyclopentyl. In some embodiments, R is optionally substituted cyclobutyl. In some embodiments, R is cyclobutyl. In some embodiments, R is optionally substituted cyclopropyl. In some embodiments, R is cyclopropyl.
In some embodiments, when R is or includes a ring structure, such as a cycloaliphatic, heterocycloaliphatic, aryl, heteroaryl, and the like, the ring structure can be monocyclic, bicyclic, or polycyclic. In some embodiments, R is or comprises a monocyclic structure. In some embodiments, R is or includes a bicyclic structure. In some embodiments, R is or comprises a polycyclic structure.
In some embodiments, R is a group having 1 to 10 substituents independently selected from oxygen, nitrogen, sulfur, phosphorus, andoptionally substituted C of hetero atoms of silicon1-30A heteroaliphatic group. In some embodiments, R is optionally substituted C having 1 to 10 heteroatoms1-20A heteroaliphatic group. In some embodiments, R is optionally substituted C having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, or silicon1-20A heteroaliphatic group, optionally comprising one or more oxidized forms of nitrogen, sulfur, phosphorus, or selenium. In some embodiments, R is optionally substituted C comprising 1 to 10 groups independently selected from1-30Heteroaliphatic group:
Figure BDA0003526174380001581
-N=、≡N、-S-、-S(O)-、-S(O)2-、-O-、=O、
Figure BDA0003526174380001582
Figure BDA0003526174380001583
in some embodiments, R is optionally substituted C6-30And (4) an aryl group. In some embodiments, R is optionally substituted phenyl. In some embodiments, R is phenyl. In some embodiments, R is substituted phenyl.
In some embodiments, R is an optionally substituted 8-to 10-membered bicyclic saturated, partially unsaturated, or aryl ring. In some embodiments, R is an optionally substituted 8-to 10-membered bicyclic saturated ring. In some embodiments, R is an optionally substituted 8-to 10-membered bicyclic moiety unsaturated ring. In some embodiments, R is an optionally substituted 8-to 10-membered bicyclic aryl ring. In some embodiments, R is optionally substituted naphthyl.
In some embodiments, R is an optionally substituted 5-to 30-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, R is an optionally substituted 5-to 30-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, and sulfur. In some embodiments, R is an optionally substituted 5-to 30-membered heteroaryl ring having 1-5 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, R is an optionally substituted 5-to 30-membered heteroaryl ring having 1 to 5 heteroatoms independently selected from oxygen, nitrogen, and sulfur.
In some embodiments, R is an optionally substituted 5-to 6-membered monocyclic heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is a substituted 5-to 6-membered monocyclic heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an unsubstituted 5-to 6-membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5-to 6-membered monocyclic heteroaryl ring having 1 to 3 heteroatoms independently selected from nitrogen, sulfur, and oxygen. In some embodiments, R is a substituted 5-to 6-membered monocyclic heteroaryl ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an unsubstituted 5-to 6-membered monocyclic heteroaryl ring having 1-3 heteroatoms independently selected from nitrogen, sulfur, and oxygen.
In some embodiments, R is an optionally substituted 5-membered monocyclic heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, R is an optionally substituted 6-membered monocyclic heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, R is an optionally substituted 5-membered monocyclic heteroaryl ring having one heteroatom selected from nitrogen, oxygen, and sulfur. In some embodiments, R is optionally substituted pyrrolyl, furanyl, or thienyl.
In some embodiments, R is an optionally substituted 5-membered heteroaryl ring having two heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 5-membered heteroaryl ring having one nitrogen atom and an additional heteroatom selected from sulfur or oxygen. In some embodiments, R is an optionally substituted 5-membered heteroaryl ring having three heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5-membered heteroaryl ring having four heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, R is an optionally substituted 6-membered heteroaryl ring having 1 to 4 nitrogen atoms. In some embodiments, R is an optionally substituted 6-membered heteroaryl ring having 1 to 3 nitrogen atoms. In other embodiments, R is an optionally substituted 6-membered heteroaryl ring having 1 to 2 nitrogen atoms. In some embodiments, R is an optionally substituted 6-membered heteroaryl ring having four nitrogen atoms. In some embodiments, R is an optionally substituted 6-membered heteroaryl ring having three nitrogen atoms. In some embodiments, R is an optionally substituted 6-membered heteroaryl ring having two nitrogen atoms. In certain embodiments, R is an optionally substituted 6-membered heteroaryl ring having one nitrogen atom.
In certain embodiments, R is an optionally substituted 8-to 10-membered bicyclic heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5, 6-fused heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 6, 6-fused heteroaryl ring having 1 to 4 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, R is a 3-to 30-membered heterocyclic ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, R is a 3 to 30 membered heterocyclic ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, and sulfur. In some embodiments, R is a 3-to 30-membered heterocyclic ring having 1-to 5 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, R is a 3 to 30 membered heterocyclic ring having 1 to 5 heteroatoms independently selected from oxygen, nitrogen, and sulfur.
In some embodiments, R is an optionally substituted 3-to 7-membered saturated or partially unsaturated heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is a substituted 3-to 7-membered saturated or partially unsaturated heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an unsubstituted 3-to 7-membered saturated or partially unsaturated heterocyclic ring having 1-3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 5-to 7-membered partially unsaturated monocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 5-to 6-membered partially unsaturated monocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 5-membered partially unsaturated monocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 6-membered partially unsaturated monocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 7-membered partially unsaturated monocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 3-membered heterocyclic ring having one heteroatom selected from nitrogen, oxygen, or sulfur. In some embodiments, R is an optionally substituted 4-membered heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5-membered heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 6-membered heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 7-membered heterocyclic ring having 1 to 3 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, R is an optionally substituted 3-membered saturated or partially unsaturated heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 4-membered saturated or partially unsaturated heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5-membered saturated or partially unsaturated heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 6-membered saturated or partially unsaturated heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 7-membered saturated or partially unsaturated heterocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is an optionally substituted 5-to 6-membered partially unsaturated monocyclic ring having 1 to 2 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In certain embodiments, R is optionally substituted tetrahydropyridinyl, dihydrothiazolyl, dihydrooxazolyl, or oxazolinyl.
In some embodiments, R is an optionally substituted 7-to 10-membered bicyclic saturated or partially unsaturated heterocyclic ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is optionally substituted indolinyl. In some embodiments, R is optionally substituted isoindolinyl. In some embodiments, R is an optionally substituted 1, 2, 3, 4-tetrahydroquinolinyl. In some embodiments, R is optionally substituted 1, 2, 3, 4-tetrahydroisoquinolinyl. In some embodiments, R is optionally substituted azabicyclo [3.2.1] octyl.
In some embodiments, R is an optionally substituted 8-to 10-membered bicyclic heteroaryl ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur. In some embodiments, R is an optionally substituted 5, 6-fused heteroaryl ring having 1 to 5 heteroatoms independently selected from nitrogen, oxygen, and sulfur.
In some embodiments, R is optionally substituted C6-30An arylaliphatic group. In some embodiments, R is optionally substituted C6-20An arylaliphatic group. In some embodiments, R is optionally substituted C6-10An arylaliphatic group. In some embodiments, the aryl portion of the arylaliphatic has 6, 10, or 14 aryl carbon atoms. In some embodiments, the aryl portion of the arylaliphatic has 6 aryl carbon atoms. In some embodiments, the aryl portion of the arylaliphatic group has 10 aryl carbon atoms. In some embodiments, the aryl portion of the arylaliphatic group has 14 aryl carbon atoms. In some embodiments, the aryl moiety is optionally substituted phenyl.
In some embodiments, R is an optionally substituted C having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group. In some embodiments, R is a heteroatom having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, and sulfur Optionally substituted C of6-30An aryl heteroaliphatic group. In some embodiments, R is an optionally substituted C having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-20An aryl heteroaliphatic group. In some embodiments, R is optionally substituted C having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, and sulfur6-20An aryl heteroaliphatic group. In some embodiments, R is optionally substituted C having 1 to 5 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-10An aryl heteroaliphatic group. In some embodiments, R is optionally substituted C having 1 to 5 heteroatoms independently selected from oxygen, nitrogen, and sulfur6-10An aryl heteroaliphatic group.
In some embodiments, two R groups optionally and independently form a covalent bond together. In some embodiments, -C ═ O is formed. In some embodiments, -C ═ C-is formed. In some embodiments, -C ≡ C-is formed.
In some embodiments, two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon in addition to the atom. In some embodiments, two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 20-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon in addition to the atom. In some embodiments, two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 10-membered monocyclic, bicyclic, or polycyclic ring having 0 to 5 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon in addition to the atom. In some embodiments, two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 6-membered monocyclic, bicyclic, or polycyclic ring having 0 to 3 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon in addition to the atom. In some embodiments, two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 5-membered monocyclic, bicyclic, or polycyclic ring having 0 to 3 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon in addition to the atom.
In some embodiments, two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 20-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 10-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 10-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 5 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 6-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 3 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon. In some embodiments, two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 5-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 3 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
In some embodiments, the heteroatoms in the R groups or in the structures formed by two or more R groups together are selected from oxygen, nitrogen, and sulfur. In some embodiments, the formed ring is 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 membered. In some embodiments, the formed ring is saturated. In some embodiments, the formed ring is partially saturated. In some embodiments, the ring formed is aromatic. In some embodiments, the formed ring comprises a saturated, partially saturated, or aromatic ring moiety. In some embodiments, the ring formed comprises 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 aromatic ring atoms. In some embodiments, the formed contains no more than 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 aromatic ring atoms. In some embodiments, the aromatic ring atoms are selected from carbon, nitrogen, oxygen, and sulfur.
In some embodiments, the ring formed by two or more R groups (or two or more groups selected from R and variables that can be R) taken together is C3-30A cycloaliphatic group; c 6-30An aryl group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; or a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon, such as the rings described for R, but which is divalent or polyvalent.
Exemplary compounds are set forth in table 1 below.
TABLE 1 exemplary Compounds
Figure BDA0003526174380001631
Figure BDA0003526174380001641
Figure BDA0003526174380001651
Figure BDA0003526174380001661
Figure BDA0003526174380001671
Figure BDA0003526174380001681
Figure BDA0003526174380001691
Figure BDA0003526174380001701
Figure BDA0003526174380001711
Figure BDA0003526174380001721
Figure BDA0003526174380001731
Figure BDA0003526174380001741
Figure BDA0003526174380001751
Figure BDA0003526174380001761
Figure BDA0003526174380001771
Figure BDA0003526174380001781
Figure BDA0003526174380001791
Figure BDA0003526174380001801
Figure BDA0003526174380001811
In some embodiments, the present disclosure provides a compound set forth in table 1 above, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-1, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-2, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-3, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-4 or a pharmaceutically acceptable salt thereof. In some embodiments, the present invention provides compound I-5 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-6 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-7, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-8, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-9, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-10, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-11, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-12, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-13, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-14 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-15, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-16, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-17 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-18, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-19, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-24, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-25, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-26, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-27, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-28, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-29, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-30, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-31 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-32, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-33, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-34, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-35, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-36, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-37 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-38, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-39, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-40, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-41, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-42, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-43, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-44, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-45, or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-46 or a pharmaceutically acceptable salt thereof. In some embodiments, the present disclosure provides compound I-47, or a pharmaceutically acceptable salt thereof.
4. General methods for providing the Compounds of the invention
The compounds of the present disclosure may generally be prepared or isolated by synthetic and/or semi-synthetic methods known to those of skill in the art for analogous compounds and by the methods described in detail in the examples herein.
In some embodiments, where particular protecting groups ("PG"), leaving groups ("LG"), or conversion conditions are depicted, one of ordinary skill in the art will appreciate that other protecting groups, leaving groups, and conversion conditions are also suitable and encompassed. The groups and transformations are described in detail in the machester advanced organic chemistry: reactions, Mechanisms and structures (March's Advanced Organic Chemistry: Reactions, mechanics, and Structure), M.B.Smith and J.March, 5 th edition, John Willi-parent-child publishing, 2001; comprehensive Organic Transformations (Comprehensive Organic Transformations), r.c. larock, 2 nd edition, john wiley parent-child publishing company, 1999; and Protecting Groups in Organic Synthesis (Protecting Groups in Organic Synthesis), t.w.greene and p.g.m.wuts, 3 rd edition, john wily parent-son publishing company, 1999, the entire contents of each of which are hereby incorporated by reference herein.
In some embodiments, the leaving group comprises, but is not limited to, a halogen (e.g., fluoride, chloride, bromide, iodide), a sulfonate (e.g., methanesulfonate, toluenesulfonate, benzenesulfonate, bromobenzenesulfonate, nitrobenzenesulfonate, trifluoromethanesulfonate), diazonium, and the like.
In some embodiments, the oxygen protecting group comprises, for example, a carbonyl protecting group, a hydroxyl protecting group, and the like. Hydroxy protecting groups are well known in the art and include protecting groups described in detail in organic synthesis, hydroxy protecting groups in t.w.greene and p.g.m.wuts, 3 rd edition, john wiley parent publishing company, 1999, the entire contents of which are incorporated herein by reference. Examples of suitable hydroxyl protecting groups include, but are not limited to, esters, allyl ethers, silyl ethers, alkyl ethers, arylalkyl ethers, and alkoxyalkyl ethers. Examples of such esters include formates, acetates, carbonates, and sulfonates. Specific examples include formates, benzoylformates, chloroacetates, trifluoroacetates, methoxyacetates, triphenylmethoxyacetates, p-chlorophenoxyacetates, 3-phenylpropionates, 4-oxopentanoates, 4- (ethylenedithio) pentanoates, pivaloates (trimethylacetyl esters), crotonates, 4-methoxy-crotonates, benzoates, p-benzylbenzoates, 2, 4, 6-trimethylbenzoates, carbonates, such as methyl esters, 9-fluorenylmethyl esters, ethyl esters, 2, 2, 2-trichloroethyl esters, 2- (trimethylsilyl) ethyl esters, 2- (phenylsulfonyl) ethyl esters, vinyl esters, allyl esters, and p-nitrobenzyl esters. Examples of the silyl ethers include trimethylsilyl ether, triethylsilyl ether, t-butyldimethylsilyl ether, t-butyldiphenylsilyl ether, triisopropylsilyl ether and other trialkylsilyl ethers. The alkyl ethers include methyl ether, benzyl ether, p-methoxybenzyl ether, 3, 4-dimethoxybenzyl ether, trityl ether, tert-butyl ether, allyl ether and allyloxycarbonyl ether or derivatives. Alkoxyalkyl ethers include acetals such as methoxymethyl ether, methylthiomethyl ether, (2-methoxyethoxy) methyl ether, benzyloxymethyl ether, β - (trimethylsilyl) ethoxymethyl ether, and tetrahydropyranyl ether. Examples of arylalkyl ethers include benzyl ether, p-methoxybenzyl ether (MPM), 3, 4-dimethoxybenzyl ether, O-nitrobenzyl ether, p-halophenyl methyl ether, 2, 6-dichlorobenzyl ether, p-cyanobenzyl ether, and 2-and 4-picolyl ethers.
Amino protecting groups are well known in the art and include protecting groups described in detail in organic synthesis, hydroxyl protecting groups in t.w.greene and p.g.m.wuts, 3 rd edition, john wiley parent publishing, 1999, the entire contents of which are incorporated herein by reference. Suitable amino protecting groups include, but are not limited to: aralkyl amines, carbamates, cyclic imides, allyl amines, amides, and the like. Examples of such groups include tert-Butoxycarbonyl (BOC), ethoxycarbonyl, methoxycarbonyl, trichloroethoxycarbonyl, allyloxycarbonyl (Alloc), benzyloxycarbonyl (CBZ), allyl, phthalimide, benzyl (Bn), fluorenylmethylcarbonyl (Fmoc), formyl, acetyl, chloroacetyl, dichloroacetyl, trichloroacetyl, phenethyl, trifluoroacetyl, benzoyl and the like.
Those skilled in the art will appreciate that compounds of formula I, II or III may contain one or more stereocenters and may exist as racemic or diastereomeric mixtures. It will also be appreciated by those skilled in the art that there are many methods known in the art for separating isomers to obtain stereoenriched or stereopure isomers of those compounds, including, but not limited to, HPLC, chiral HPLC, fractional crystallization of diastereomeric salts, enzymatic kinetic resolution (e.g., by fungal, bacterial or animal derived lipases or esterases) and the use of enantiomeric enrichment reagents to form covalent diastereomeric derivatives.
It will be appreciated by those skilled in the art that the various functional groups (e.g., aliphatic groups, alcohols, carboxylic acids, esters, amides, aldehydes, halogens, and nitriles) present in the compounds of the present disclosure can be interconverted by techniques well known in the art, including, but not limited to, reduction, oxidation, esterification, hydrolysis, partial oxidation, partial reduction, halogenation, dehydration, partial hydration, and hydration. "organic chemistry high horse, 5 th edition, editor: smith, m.b. and March, j., john willi parent-child publishing company, new york: 2001, the entire contents of which are incorporated herein by reference. The interconversion may require one or more of the foregoing techniques, and certain methods for synthesizing the compounds of the present disclosure are described in the examples below.
In some embodiments, the present disclosure provides compounds useful for the preparation of ARM. In some embodiments, the present disclosure provides compounds suitable for use in the construction of ARM molecules via cycloaddition reactions (e.g., click chemistry or variants thereof).
In some embodiments, the present disclosure provides a compound having the structure of formula IV:
Figure BDA0003526174380001841
or a salt thereof, wherein
ABT is an antibody binding moiety;
l is a linker moiety;
RdIs or includes a reactive group;
each LaIndependently a covalent bond, or is selected from C1-C20Aliphatic radical or C having 1 to 5 hetero atoms1-C20A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from: c3-20A cycloaliphatic ring; c6-20An aryl ring; a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3 to 20 membered heterocyclyl ring having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or optionally substituted selected fromGroup (b): c1-30An aliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon1-30A heteroaliphatic group; c6-30An aryl group; c6-30An arylaliphatic group; c having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon6-30An aryl heteroaliphatic group; a 5-to 30-membered heteroaryl having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; and a 3-to 30-membered heterocyclic group having 1 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus and silicon, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the atom, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having, in addition to the intervening atoms, 0 to 10 heteroatoms independently selected from oxygen, nitrogen, sulfur, phosphorus, and silicon.
As appreciated by those skilled in the art and demonstrated herein, a variety of reactive groups can be utilized in accordance with the present disclosure. For example, in some embodiments, Rdis-La-R', wherein Rdcomprising-C ≡ C-or-N3(ii) a Such RdIt is especially suitable for click chemistry reaction.
In some embodiments, the present disclosure provides a compound of formula IV-a:
Figure BDA0003526174380001851
or a salt thereof, wherein each variable is independently as described in the disclosure.
In some embodiments, the present disclosure provides a compound of formula IV-b:
Rc-(Xaa)z-L-Rd
IV-b
or a salt thereof, wherein each variable is independently as described in the disclosure.
In some embodiments, the present disclosure provides a compound of formula IV-c:
Figure BDA0003526174380001852
or a salt thereof, wherein each variable is independently as described in the disclosure.
In some embodiments, the present disclosure provides a compound of formula IV-d:
Figure BDA0003526174380001861
or a salt thereof, wherein each variable is independently as described in the disclosure.
In some embodiments, the present disclosure provides a compound of formula V:
Figure BDA0003526174380001862
or a salt thereof, wherein each variable is independently as described in the disclosure.
In some embodiments of the present invention, the,
Figure BDA0003526174380001863
as described herein
Figure BDA0003526174380001864
In some embodiments, the target binding moiety binds to CD 38.
In some embodiments, the present disclosure provides a method comprising:
a) providing a first compound comprising a target binding moiety as described herein and a first reactive group;
b) providing a second compound comprising an antibody binding moiety as described herein and a second reactive group; and
c) reacting the first reactive group with the second reactive group to covalently link the target binding moiety to the antibody binding moiety.
In some embodiments, the first compound is a compound of formula IV, IV-a, IV-b, IV-c, or IV-d, or a salt thereof. In some embodiments, the second compound is a compound of formula V or a salt thereof.
In some embodiments, the present disclosure provides a method of preparing a compound, comprising the steps of:
providing a first compound having formula IV, IV-a, IV-b, IV-c, or IV-d, or a salt thereof, wherein the compound comprises a first reactive moiety;
providing a second compound having formula V or a salt thereof comprising a second reactive moiety; and
reacting the first compound with the second compound, wherein the first reactive moiety reacts with the second reactive moiety to link the first compound and the second compound.
Various reactive moieties and reactions can be utilized in accordance with the present disclosure. For example, in some embodiments, the reaction is an amidation reaction, wherein the first reactive moiety (e.g., R)d) And a second reactive moiety (e.g., R)d) Is or includes an activated carboxylic acid (e.g., is or includes
Figure BDA0003526174380001871
) And the other is or includes an amino group.
In some embodiments, the present disclosure provides a method of preparing a compound, comprising the steps of:
providing a first compound having formula IV, IV-a, IV-b, IV-c, or IV-d, or a salt thereof, wherein the compound comprises a first reactive moiety;
providing a second compound having formula V or a salt thereof comprising a second reactive moiety; and
Reacting the first compound with the second compound, wherein the first reactive moiety reacts with the second reactive moiety via a cycloaddition reaction.
A number of cycloaddition reactions may be utilized in accordance with the present disclosure. In some embodiments, the cycloaddition reaction is [4+2 ]]And (4) reacting. In some embodiments, the cycloaddition reaction is [3+2 ]]And (4) reacting. In some embodiments, [3+2 ]]The reaction is a click chemistry reaction. In some embodiments, the first reactive moiety is-C ≡ C-and the second reactive moiety is-N3. In some embodiments, the first reactive moiety is-N3And the second reactive moiety is-C ≡ C-.
In some embodiments, provided techniques (e.g., compounds, agents, etc.) are or include peptide moieties. Those skilled in the art will appreciate that variable peptide synthesis techniques are readily available and can be utilized in accordance with the present disclosure.
Various techniques are available and can be used to assess provided techniques, such as the properties and/or activities of provided compounds (e.g., CD38 binding, immune activity recruitment, ADCC, etc.), in accordance with the present disclosure. Certain useful techniques are described in the examples.
5. Use, formulation and application
Pharmaceutically acceptable compositions
According to another embodiment, the present disclosure provides a composition comprising a compound described herein, or a pharmaceutically acceptable derivative thereof, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In some embodiments, the present disclosure provides a pharmaceutical composition comprising a compound of the present disclosure (e.g., ARM) and a pharmaceutically acceptable carrier. In some embodiments, the present disclosure provides a pharmaceutical composition comprising a therapeutically effective amount of a compound of the present disclosure (e.g., ARM) and a pharmaceutically acceptable carrier. In some embodiments, the amount of the compound in the composition is such that it is effective to selectively direct the antibody to a target, such as a diseased cell (e.g., a cancer cell), and/or to induce an antibody-directed activity, such as cell-mediated immunity, e.g., cytotoxicity. In certain embodiments, the amount of the compound in the composition is such that it is effective to selectively direct the antibody to cancer cells expressing CD38 in a biological sample or in a subject (e.g., a cancer patient) and induce antibody-directed activity, such as cell-mediated cytotoxicity. In certain embodiments, the compositions are formulated for administration to a patient in need of such compositions. In some embodiments, the composition is formulated for oral administration to a patient.
In some embodiments, a pharmaceutically acceptable carrier, adjuvant, or vehicle is a non-toxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants, or vehicles may include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins (e.g., human serum albumin), buffer substances (e.g., phosphates), glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts, or electrolytes (e.g., protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts), colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene-polyoxypropylene block polymers, polyethylene glycol, and lanolin.
In some embodiments, a pharmaceutically acceptable derivative is a non-toxic salt, ester, salt of an ester, or other derivative of a compound that is capable of providing, directly or indirectly, the compound or an active metabolite or residue thereof upon administration to a recipient.
The compositions may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally, or via an implantable reservoir. In some embodiments, parenteral administration comprises subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. In some embodiments, the composition is administered orally, intraperitoneally, or intravenously. Sterile injectable forms of the compositions can be aqueous or oleaginous suspensions. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that may be employed are water, ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
In some embodiments, mild fixed oils, including synthetic mono-or diglycerides, may be employed. Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils (e.g., olive oil or castor oil, especially in their polyoxyethylated versions). These oil solutions or suspensions may also contain a long chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersants commonly used in formulating pharmaceutically acceptable dosage forms, including emulsions and suspensions. Other commonly used surfactants such as Tween, Span and other emulsifiers or bioavailability enhancers commonly used in the manufacture of pharmaceutically acceptable solid, liquid or other dosage forms may also be used for formulation purposes.
The pharmaceutically acceptable composition may be administered orally in any orally acceptable dosage form, including but not limited to capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in capsule form, suitable diluents include lactose and dried corn starch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
In some embodiments, the pharmaceutically acceptable composition may be administered in the form of suppositories for rectal administration. In some embodiments, these suppositories may be prepared by mixing the agent with a suitable non-irritating excipient which is solid at room temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
In some embodiments, the pharmaceutically acceptable composition may also be administered topically, particularly when the target of treatment comprises an area or organ readily accessible by topical administration, including diseases of the eye, skin, or lower intestinal tract. Topical formulations suitable for each of these areas or organs are readily prepared.
Topical administration to the lower intestinal tract may be achieved in rectal suppository formulations (see above) or in suitable enema formulations. Topical transdermal patches may also be used.
For topical administration, pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active ingredient suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of the present disclosure include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active ingredient suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
For ophthalmic use, the pharmaceutically acceptable composition may be formulated as a micronized suspension or preferably as a solution in pH adjusted isotonic sterile saline, with or without a preservative such as benzalkonium chloride. Alternatively, for ophthalmic use, the pharmaceutically acceptable composition may be formulated in an ointment such as petrolatum.
The pharmaceutically acceptable composition may also be administered by nasal spray or inhalation. Such compositions are prepared according to techniques well known in the art of pharmaceutical formulation and may be prepared as solutions in physiological saline using benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons and/or other conventional solubilizing or dispersing agents.
In some embodiments, the pharmaceutically acceptable composition is formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, the pharmaceutically acceptable composition is not administered with food. In other embodiments, the pharmaceutically acceptable composition is administered with food.
The amount of compound that can be combined with the carrier material to produce a composition in a single dosage form will vary depending on the host treated, the particular mode of administration. In some embodiments, the provided compositions are formulated such that a patient receiving these compositions can be administered a dose of between 0.01 to 100mg per kg body weight per day of the inhibitor.
It will also be understood that the specific dose and treatment regimen for any particular patient will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present disclosure in a composition will also depend on the particular compound in the composition.
Use of compounds and pharmaceutically acceptable compositions
The compounds and compositions described herein are generally useful for selectively directing antibodies to a target, in particular a target that expresses or includes CD38, such as a diseased cell (e.g., a cancer cell), and/or for inducing antibody-directed activities, such as a cell-mediated immune response (e.g., cytotoxicity).
In some embodiments, the present disclosure provides methods of recruiting an antibody (e.g., an endogenous antibody) to a target, comprising contacting the target with a provided agent, compound, or composition. In some embodiments, the recruited antibodies comprise one or more endogenous antibodies. In some embodiments, the recruited antibody is specific for one or more antigens. In some embodiments, the recruited antibody is specific for one or more peptide antigens or proteins. In some embodiments, the recruited antibody is heterogeneous in that it is not an antibody to the same antigen or protein.
In some embodiments, the present disclosure provides methods of recruiting immune cells to a target, comprising contacting the target with a provided agent, compound, or composition.
In some embodiments, the present disclosure provides methods for triggering, generating, promoting, and/or enhancing one or more immune system activities against a target comprising contacting the target with a provided agent, compound, or composition. In some embodiments, the immune system activity is or comprises ADCC. In some embodiments, the immune system activity is or comprises ADCP. In some embodiments, the immune system activity is or includes both ADCC and ADCP. In some embodiments, the immune system activity is or comprises Complement Dependent Cytotoxicity (CDC). In some embodiments, the immune system activity is or comprises ADCVI.
In some embodiments, the target is a cancer cell. In some embodiments, the target is a cancer cell in the subject. In some embodiments, the provided methods comprise administering the provided agents, compounds, or compositions to a subject.
In some embodiments, provided agents and compounds form complexes with antibodies and Fc receptors on target cells when contacted with their targets. In some embodiments, the present disclosure provides a composite comprising:
A medicament, comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
the optional presence of a linker moiety or moieties,
an Fc region, and
an Fc receptor.
In some embodiments, the antibody binding moiety is a universal antibody binding moiety. In some embodiments, the target binding moiety may bind to CD 38.
In some embodiments, the present disclosure provides a composite comprising two or more composites, each independently comprising:
a medicament, comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
the optional presence of a linker moiety or moieties,
an Fc region, and
an Fc receptor for a protein having a high Fc activity,
wherein the Fc region of the complex is that of an antibody and/or fragment thereof directed against a different antigen or protein.
In some embodiments, the Fc region of the complex is an Fc region of an antibody and/or fragment thereof directed to a different protein. In some embodiments, the one or more Fc regions are Fc regions of endogenous antibodies and/or fragments thereof.
In some embodiments, the present disclosure provides a method of treating one or more disorders, diseases, and/or conditions, wherein the disorder, disease, or condition is cancer.
In some embodiments, "neoplasia" or "cancer" is or includes the pathological process that results in the formation and growth of a cancerous or malignant neoplasm, i.e., abnormal tissue that grows by cell proliferation, is often faster than normal tissue, and continues to grow after cessation of the stimulus that elicits new growth. Malignant neoplasms exhibit a partial or complete lack of structural organization and function coordinated with normal tissue and most invasive surrounding tissues, metastasize to several sites, and may recur after attempted removal and cause patient death unless adequately treated. As used herein, the term neoplasia is used to describe all cancerous disease conditions and encompasses or encompasses pathological processes associated with malignant hematopoiesis, ascites, and solid tumors. Representative cancers include, for example, prostate cancer, metastatic prostate cancer, stomach cancer, colon cancer, rectal cancer, liver cancer, pancreatic cancer, lung cancer, breast cancer, cervical cancer, uterine corpus cancer, ovarian cancer, testicular cancer, bladder cancer, kidney cancer, brain/CNS cancer, head and neck cancer, throat cancer, hodgkin's disease, non-hodgkin's lymphoma, multiple myeloma, leukemia, melanoma, non-melanoma skin cancer, acute lymphocytic leukemia, acute myelogenous leukemia, Ewing's sarcoma, small cell lung cancer, choriocarcinoma, rhabdomyosarcoma, Wilms' tumor, neuroblastoma, hairy cell leukemia, oral/pharyngeal cancer, esophageal cancer, laryngeal cancer, kidney cancer, and lymphoma, among others, that can be treated by one or more compounds according to the present disclosure. Furthermore, the provided techniques (e.g., compounds, compositions, methods, etc.) are particularly useful for preventing and/or treating cancer.
Furthermore, the present disclosure provides the use of a compound according to the definitions herein or a pharmaceutically acceptable salt or hydrate or solvate thereof in the manufacture of a medicament for the treatment of a proliferative disease.
In some embodiments, the present disclosure provides techniques, e.g., agents, compounds (e.g., ARM), compositions, methods, etc., that are particularly useful for treating CD 38-associated cancers. In some embodiments, the provided techniques are particularly useful for treating cancers that express or include CD38 by cancer cells.
Combination therapy
In some embodiments, the provided techniques are administered with one or more additional therapeutic agents and/or techniques (e.g., for cancer, one or more of additional therapeutic agents, surgery, radiotherapy, etc.). In some embodiments, additional therapeutic agents and/or techniques suitable for combination are those that have been used to treat the condition, disorder or disease. In certain embodiments, a provided compound (e.g., ARM) or composition thereof is administered in combination with another therapeutic agent.
Examples of agents for combination include (but are not limited to): for the treatment of Alzheimer's Disease, e.g. Alzheimer's Disease
Figure BDA0003526174380001921
And
Figure BDA0003526174380001922
for the treatment of HIV, such as ritonavir (ritonavir); for the treatment of Parkinson's Disease, such as L-DOPA/carbidopa (carbidopa), entacapone (entacapone), ropinirole (roprole), pramipexole (pramipexole), bromocriptine (bromocriptine), pergolide (pergolide), trihexyphenidyl (trihexyphenyl) and amantadine; agents for the treatment of Multiple Sclerosis (MS), e.g. interferon beta (e.g. interferon beta)
Figure BDA0003526174380001923
And
Figure BDA0003526174380001924
)、
Figure BDA0003526174380001925
and mitoxantrone (mitoxantrone); for treatment of asthma, e.g. albuterol and
Figure BDA0003526174380001926
agents for treating schizophrenia, such as repulper (zyprexa), risperidone (risperdal), selazen (seroquel) and haloperidol (haloperidol); anti-inflammatory agents, such as corticosteroids, TNF blockers, IL-1 RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents, such as cyclosporine, tacrolimus (tacrolimus), rapamycin (rapamycin), mycophenolate mofetil, interferons, corticosteroids, cyclophosphamide, azathioprine, and sulfasalazine; neurotrophic factors such as acetylcholinesterase inhibitors, MAO inhibitors, interferons, anticonvulsants, ion channel blockers, riluzole (riluzole), and antiparkinson agents; agents for the treatment of cardiovascular diseases, such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers and statins; agents for treating liver diseases, such as corticosteroids, cholestyramine, interferons, and antiviral agents; agents for treating blood disorders, such as corticosteroids, anti-leukemic agents, and growth factors; agents that prolong or improve pharmacokinetics, such as cytochrome P450 inhibitors (i.e., inhibitors of metabolic breakdown) and CYP3a4 inhibitors (e.g., ketenozole and ritonavir); and agents for treating immunodeficiency disorders, such as gamma globulin.
In certain embodiments, the provided compounds or compositions thereof are used in combination with a monoclonal antibody or siRNA therapeutic.
The additional agents can be administered separately from the provided compounds or compositions. Alternatively, the additional agent may be part of a single dosage form, mixed in a single composition with the provided compound. If administered as part of a multiple dosing regimen, the active agents may be administered simultaneously, sequentially or separated from each other by a period of time (typically within five hours of each other).
Combination therapy may include the simultaneous or sequential administration of therapeutic agents according to the present disclosure. For example, the combination may be administered simultaneously or sequentially in separate unit dosage forms, or together in a single unit dosage form.
In some embodiments, the amount of additional therapeutic agent does not exceed the amount that would be administered when the additional therapeutic agent is not combined with a compound or composition provided herein.
In one embodiment, the present disclosure provides a composition comprising a compound of formula I, II or III and one or more additional therapeutic agents. The therapeutic agent may be administered with a compound of formula I, II or III, or may be administered before or after administration of a compound of formula I, II or III. Suitable therapeutic agents are described in more detail below. In certain embodiments, a compound of formula I, II or III can be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours prior to the therapeutic agent. In other embodiments, a compound of formula I, II or III can be administered up to 5 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours, 13 hours, 14 hours, 15 hours, 16 hours, 17 hours, or 18 hours after the therapeutic agent.
In some embodiments, the present disclosure provides a method of treating an inflammatory disease, disorder, or condition by administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents. Such additional therapeutic agents may be small molecules or recombinant biological agents and include, for example: an acetamidophenol; nonsteroidal anti-inflammatory drugs (NSAIDS), such as aspirin (aspirin)) Ibuprofen (ibuprofen), naproxen (naproxen), etodolac (etodolac)
Figure BDA0003526174380001931
And celecoxib (celecoxib); colchicine (colchicine)
Figure BDA0003526174380001932
Corticosteroids such as prednisone (prednisone), prednisolone (prednisone), methylprednisolone, hydrocortisone (hydrocortisone), and the like; probenecid (probenecid); allopurinol (allopurinol); febuxostat (febuxostat)
Figure BDA0003526174380001933
Sulfasalazine
Figure BDA0003526174380001934
Antimalarial drugs, e.g. hydroxychloroquine
Figure BDA0003526174380001935
And chloroquine
Figure BDA0003526174380001936
Methotrexate (methotrexate)
Figure BDA0003526174380001937
Gold salts, e.g. gold thioglucoside
Figure BDA0003526174380001938
Gold thiomalate
Figure BDA0003526174380001939
And auranofin (auranofin)
Figure BDA00035261743800019310
D-penicillamine (A)
Figure BDA00035261743800019311
Or
Figure BDA00035261743800019312
) (ii) a Azathioprine
Figure BDA00035261743800019313
Cyclophosphamide
Figure BDA00035261743800019314
Chlorambucil
Figure BDA00035261743800019315
Ciclosporin
Figure BDA00035261743800019316
Leflunomide (leflunomide)
Figure BDA00035261743800019317
And "anti-TNF" agents, such as etanercept (etanercept)
Figure BDA00035261743800019318
Infliximab (infliximab)
Figure BDA00035261743800019319
Golimumab (golimumab)
Figure BDA00035261743800019320
Cetuzumab ozolomide (certolizumab pegol)
Figure BDA00035261743800019321
And adalimumab (adalimumab)
Figure BDA00035261743800019322
"anti-IL-1" agents, e.g. anakinra
Figure BDA00035261743800019323
Heirancicept (rilonacept)
Figure BDA00035261743800019324
Kana monoclonal antibody (canakinumab)
Figure BDA00035261743800019325
anti-Jak inhibitors, such as tofacitinib; antibodies, e.g. rituximab (rituximab)
Figure BDA00035261743800019326
"anti-T cell" agents, e.g. acappe (abatacept)
Figure BDA00035261743800019327
"anti-IL-6" agents, e.g. tositumumab (tocilizumab)
Figure BDA00035261743800019328
Diclofenac (diclofenac); cortisone; hyaluronic acid (A)
Figure BDA00035261743800019329
Or
Figure BDA00035261743800019330
) (ii) a Monoclonal antibodies, such as, for example, tanizumab; anticoagulants, e.g. heparin (A), (B)
Figure BDA0003526174380001941
Or
Figure BDA0003526174380001942
) And warfarin (warfarin)
Figure BDA0003526174380001943
Antidiarrheals, e.g. diphenoxylate
Figure BDA0003526174380001944
And loperamide (loperamide)
Figure BDA0003526174380001945
Bile acid binders such as cholestyramine; alosetron (alosetron)
Figure BDA0003526174380001946
Lubiprostone(1ubiprostone)
Figure BDA0003526174380001947
Laxatives, e.g. magnesium milk, polyethylene glycol
Figure BDA0003526174380001948
And
Figure BDA0003526174380001949
anticholinergic or antispasmodic agents, e.g. bicyclic amines
Figure BDA00035261743800019410
Beta-2 agonists, e.g. salbutamol(s) ((R))
Figure BDA00035261743800019411
HFA、
Figure BDA00035261743800019412
HFA), levalbuterol
Figure BDA00035261743800019413
Ocinalin (metaprotenol)
Figure BDA00035261743800019414
Pibuterol acetate (pirbuterol acetate)
Figure BDA00035261743800019415
Terbutaline sulfate (terbutaline sulfate)
Figure BDA00035261743800019416
Salmeterol xinafoate (salmeterol)
Figure BDA00035261743800019417
And formoterol (formoterol)
Figure BDA00035261743800019418
Anticholinergic agents, for example ipratropium bromide
Figure BDA00035261743800019419
And tiotropium (tiotropium)
Figure BDA00035261743800019420
Inhaled corticosteroids, such as beclomethasone dipropionate (diproprionate) (Beclomethione)
Figure BDA00035261743800019421
And
Figure BDA00035261743800019422
) Triamcinolone acetonide (triamcinolone acetonide)
Figure BDA00035261743800019423
Mometasone (mometasone)
Figure BDA00035261743800019424
Budesonide (budesonide)
Figure BDA00035261743800019425
And flunisolide (flunisolide)
Figure BDA00035261743800019426
Figure BDA00035261743800019427
Cromolyn sodium salt
Figure BDA00035261743800019428
Methylxanthines, e.g. theophylline
Figure BDA00035261743800019429
Figure BDA00035261743800019430
And aminophylline; IgE antibodies, e.g. omalizumab
Figure BDA00035261743800019431
Nucleoside reverse transcriptase inhibitors, e.g. zidovudine(s) ((R))zidovudine)
Figure BDA00035261743800019432
Abacavir (abacavir)
Figure BDA00035261743800019433
Abacavir/lamivudine (lamivudine)
Figure BDA00035261743800019434
Abacavir/lamivudine/zidovudine
Figure BDA00035261743800019435
Didanosine (didanosine)
Figure BDA00035261743800019436
Emtricitabine (emtricitabine)
Figure BDA00035261743800019437
Lamivudine
Figure BDA00035261743800019438
Lamivudine/zidovudine
Figure BDA00035261743800019439
Stavudine (stavudine)
Figure BDA00035261743800019440
And zalcitabine (zalcitabine)
Figure BDA00035261743800019441
Non-nucleoside reverse transcriptase inhibitors, e.g. delavirdine
Figure BDA00035261743800019442
Efavirenz (efavirenz)
Figure BDA00035261743800019443
Nevirapine (nevairapine)
Figure BDA00035261743800019444
And etravirine (etravirine)
Figure BDA00035261743800019445
Nucleotide reverse transcriptase inhibitors, e.g. tenofovir (tenofovir)
Figure BDA00035261743800019446
Protease inhibitors, e.g. amprenavir (amprenavir)
Figure BDA00035261743800019447
Atazanavir (atazanavir)
Figure BDA00035261743800019448
Darunavir (darunavir)
Figure BDA00035261743800019449
Fusavir (fosamprenavir)
Figure BDA00035261743800019450
Indinavir (indinavir)
Figure BDA00035261743800019451
Lopinavir (lopinavir) and ritonavir
Figure BDA00035261743800019452
Nelfinavir (nelfinavir)
Figure BDA00035261743800019453
Ritonavir
Figure BDA00035261743800019454
Saquinavir (saquinavir) ((S))
Figure BDA00035261743800019455
Or
Figure BDA00035261743800019456
) And tipranavir (tipranavir)
Figure BDA00035261743800019457
Entry inhibitors, e.g. enfuvirtide
Figure BDA00035261743800019458
And maraviroc (maravroc)
Figure BDA00035261743800019459
Integrase inhibitors, e.g. raltegravir
Figure BDA00035261743800019460
Adriamycin (doxorubicin)
Figure BDA00035261743800019461
Vincristine (vincristine)
Figure BDA00035261743800019462
Bortezomib (bortezomib)
Figure BDA00035261743800019463
And dexamethasone (dexamethasone)
Figure BDA00035261743800019464
With lenalidomide
Figure BDA0003526174380001951
A combination of (1); or any combination thereof.
In some embodiments, the present disclosure provides a method of treating gout comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from: nonsteroidal anti-inflammatory drugs (NSAIDS), e.g. aspirin, ibuprofen, naproxen, etodolac
Figure BDA0003526174380001952
And celecoxib; colchicine
Figure BDA0003526174380001953
Cortex (cortex)Steroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like; probenecid; allopurinol; and febuxostat
Figure BDA0003526174380001954
In some embodiments, the present disclosure provides a method of treating rheumatoid arthritis comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: nonsteroidal anti-inflammatory drugs (NSAIDS), e.g. aspirin, ibuprofen, naproxen, etodolac
Figure BDA0003526174380001955
And celecoxib; corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like; sulfasalazine
Figure BDA0003526174380001956
Antimalarial drugs, e.g. hydroxychloroquine
Figure BDA0003526174380001957
And chloroquine
Figure BDA0003526174380001958
Methotrexate (MTX)
Figure BDA0003526174380001959
Gold salts, e.g. gold thioglucoside
Figure BDA00035261743800019510
Gold thiomalate
Figure BDA00035261743800019511
And auranofin
Figure BDA00035261743800019512
D-penicillamine (A)
Figure BDA00035261743800019513
Or
Figure BDA00035261743800019514
) (ii) a Azathioprine
Figure BDA00035261743800019515
Cyclophosphamide
Figure BDA00035261743800019516
Chlorambucil
Figure BDA00035261743800019517
Ciclosporin
Figure BDA00035261743800019518
Leflunomide
Figure BDA00035261743800019519
And "anti-TNF" agents, e.g. etanercept
Figure BDA00035261743800019520
Infliximab
Figure BDA00035261743800019521
Gollimumab
Figure BDA00035261743800019522
Cytuzumab ozogamicin
Figure BDA00035261743800019523
And adalimumab
Figure BDA00035261743800019524
"anti-IL-1" agents, e.g. anakinra
Figure BDA00035261743800019525
And linaglicept
Figure BDA00035261743800019526
Antibodies, e.g. rituximab
Figure BDA00035261743800019527
"anti-T cell" agents, e.g. Albapup
Figure BDA00035261743800019528
And "anti-IL-6" agents, e.g. tositumumab
Figure BDA00035261743800019529
In some embodiments, the present disclosure provides a method of treating osteoarthritis comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from: an acetamidophenol; nonsteroidal anti-inflammatory drugs (NSAIDS), e.g. aspirin, ibuprofen, naproxen, etodolac
Figure BDA00035261743800019530
And celecoxib; diclofenac acid; cortisone; hyaluronic acid (A)
Figure BDA00035261743800019531
Or
Figure BDA00035261743800019532
) (ii) a And monoclonal antibodies, such as tanlizumab.
In some embodiments, the present disclosure provides a method of treating lupus, comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from: an acetamidophenol; nonsteroidal anti-inflammatory drugs (NSAIDS), e.g. aspirin, ibuprofen, naproxen, etodolac
Figure BDA00035261743800019533
And celecoxib; corticosteroids such as prednisone, prednisolone, methylprednisolone, hydrocortisone, and the like; antimalarial drugs, e.g. hydroxychloroquine
Figure BDA00035261743800019534
And chloroquine
Figure BDA00035261743800019535
Cyclophosphamide
Figure BDA00035261743800019536
Methotrexate (MTX)
Figure BDA00035261743800019537
Azathioprine
Figure BDA00035261743800019538
And anticoagulants, e.g. heparin (C)
Figure BDA00035261743800019539
Or
Figure BDA00035261743800019540
) And warfarin
Figure BDA00035261743800019541
In some embodiments, the present disclosure provides a method of treating inflammatory bowel disease comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: meishalaming (mesalamine)
Figure BDA0003526174380001961
Sulfasalazine
Figure BDA0003526174380001962
Antidiarrheals, e.g. diphenoxylate
Figure BDA0003526174380001963
And loperamide
Figure BDA0003526174380001964
Bile acid binders such as cholestyramine; alosetron
Figure BDA0003526174380001965
Lubiprostone
Figure BDA0003526174380001966
Laxatives, e.g. magnesium milk, polyethylene glycol
Figure BDA0003526174380001967
And
Figure BDA0003526174380001968
and anticholinergic or antispasmodic agents, e.g. bicyclic amines
Figure BDA0003526174380001969
anti-TNF therapy; a steroid; and antibiotics, such as metronidazole (Flagyl) or ciprofloxacin (ciprofloxacin).
In some embodiments, the present disclosure provides a method of treating asthma comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of:
Figure BDA00035261743800019610
beta-2 agonists, e.g. salbutamol(s) ((R))
Figure BDA00035261743800019611
HFA、
Figure BDA00035261743800019612
HFA), levalbuterol
Figure BDA00035261743800019613
Ocinalin
Figure BDA00035261743800019614
Pirbuterol acetate
Figure BDA00035261743800019615
Terbutaline sulfate
Figure BDA00035261743800019616
Salmeterol xinafoate
Figure BDA00035261743800019617
And formoterol
Figure BDA00035261743800019618
Anticholinergic agents, e.g. ipratropium bromide
Figure BDA00035261743800019619
And tiotropium
Figure BDA00035261743800019620
Inhaled corticosteroids, such as prednisone, prednisolone, beclomethasone dipropionate: (
Figure BDA00035261743800019621
Figure BDA00035261743800019622
And
Figure BDA00035261743800019623
) Triamcinolone acetonide
Figure BDA00035261743800019624
Mometasone
Figure BDA00035261743800019625
Budesonide
Figure BDA00035261743800019626
Fluniprole
Figure BDA00035261743800019627
And
Figure BDA00035261743800019628
cromolyn sodium salt
Figure BDA00035261743800019629
Methylxanthines, e.g. theophylline
Figure BDA00035261743800019630
And aminophylline; and IgE antibodies, e.g. OlympicMazuki single antibody
Figure BDA00035261743800019631
In some embodiments, the present disclosure provides a method of treating COPD comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from: beta-2 agonists, e.g. salbutamol(s) ((R))
Figure BDA00035261743800019632
HFA、
Figure BDA00035261743800019633
HFA), levalbuterol
Figure BDA00035261743800019634
Ocinalin
Figure BDA00035261743800019635
Pirbuterol acetate
Figure BDA00035261743800019636
Terbutaline sulfate
Figure BDA00035261743800019637
Salmeterol xinafoate
Figure BDA00035261743800019638
And formoterol
Figure BDA00035261743800019639
Anticholinergic agents, e.g. ipratropium bromide
Figure BDA00035261743800019640
And tiotropium
Figure BDA00035261743800019641
Methylxanthines, e.g. theophylline
Figure BDA00035261743800019642
And aminophylline; inhaled corticosteroids, such as prednisone, prednisolone, beclomethasone dipropionate: (
Figure BDA00035261743800019643
Figure BDA00035261743800019644
And
Figure BDA00035261743800019645
) Triamcinolone acetonide
Figure BDA00035261743800019646
Mometasone
Figure BDA00035261743800019647
Budesonide
Figure BDA00035261743800019648
Fluniprole
Figure BDA00035261743800019649
And
Figure BDA00035261743800019650
in some embodiments, the present disclosure provides a method of treating HIV comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: nucleoside reverse transcriptase inhibitors, e.g. zidovudine
Figure BDA00035261743800019651
Abacavir
Figure BDA00035261743800019652
Abacavir/lamivudine
Figure BDA00035261743800019653
Abacavir/lamivudine/zidovudine
Figure BDA00035261743800019654
Defenoxin
Figure BDA00035261743800019655
Emtricitabine
Figure BDA00035261743800019656
Lamivudine
Figure BDA00035261743800019657
Lamivudine/zidovudine
Figure BDA00035261743800019658
Stavudine
Figure BDA00035261743800019659
And zalcitabine
Figure BDA0003526174380001971
Non-nucleoside reverse transcriptase inhibitors, e.g. delavirdine
Figure BDA0003526174380001972
Efavirenz
Figure BDA0003526174380001973
Nevirapine
Figure BDA0003526174380001974
And etravirine
Figure BDA0003526174380001975
Nucleotide reverse transcriptase inhibitors, e.g. tenofovir
Figure BDA0003526174380001976
Protease inhibitors, e.g. amprenavir
Figure BDA0003526174380001977
Atazanavir
Figure BDA0003526174380001978
Darunavir
Figure BDA0003526174380001979
Fusavir
Figure BDA00035261743800019710
Indinavir
Figure BDA00035261743800019711
Lopinavir and ritonavir
Figure BDA00035261743800019712
Nelfinavir
Figure BDA00035261743800019713
Ritonavir
Figure BDA00035261743800019714
Saquinavir (a)
Figure BDA00035261743800019715
Or
Figure BDA00035261743800019716
) And tipranavir
Figure BDA00035261743800019717
Entry inhibitors, e.g. Enfuvirtide
Figure BDA00035261743800019718
And maraviroc
Figure BDA00035261743800019719
Integrase inhibitors, e.g. latiravir
Figure BDA00035261743800019720
And combinations thereof.
In some embodiments, the present disclosure provides a method of treating cancer comprising administering to a subject in need thereof a compound of formula I, II or III and one or more additional therapeutic agents. In some embodiments, the cancer is multiple myeloma. In some embodiments, the additional therapeutic agent is lenalidomide. In some embodiments, the additional therapeutic agent is bortezomib. In some embodiments, the additional therapeutic agent is dexamethasone. In some embodiments, the additional therapeutic agent is melphalan (melphalan). In some embodiments, the additional therapeutic agent is prednisone. In some embodiments, the combination is a combination of a provided compound (e.g., ARM) with lenalidomide or bortezomib and dexamethasone. In some embodiments, the combination is a combination of a provided compound (e.g., ARM) with bortezomib, melphalan, and prednisone.
In some embodiments, as demonstrated herein, the provided compounds (e.g., ARM) have low toxicity (e.g., less complement activation, less reduction of effector cells expressing CD38, etc.) compared to CD38 antibody therapy. In some embodiments, a provided compound or composition thereof, when administered as monotherapy or as part of a combination therapy, may be administered at higher and/or more frequent doses and/or for longer periods of time than other therapies (e.g., CD38 antibody therapy).
In some embodiments, the present disclosure provides a method of treating a hematologic malignancy comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from: rituximab
Figure BDA00035261743800019721
Cyclophosphamide
Figure BDA00035261743800019722
Adriamycin
Figure BDA00035261743800019723
Vincristine
Figure BDA00035261743800019724
Prednisone, hedgehog signaling inhibitors, BTK inhibitors, JAK/pan-JAK inhibitors, TYK2 inhibitors, PI3K inhibitors, SYK inhibitors, and combinations thereof.
In some embodiments, the disclosureThere is provided a method of treating a solid tumor comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: rituximab
Figure BDA00035261743800019725
Cyclophosphamide
Figure BDA00035261743800019726
Adriamycin
Figure BDA00035261743800019727
Vincristine
Figure BDA00035261743800019728
Prednisone, hedgehog signaling inhibitors, BTK inhibitors, JAK/pan-JAK inhibitors, TYK2 inhibitors, PI3K inhibitors, SYK inhibitors, and combinations thereof.
In some embodiments, the present disclosure provides a method of treating a hematologic malignancy comprising administering to a patient in need thereof a compound of formula I, II or III and a hedgehog (Hh) signaling pathway inhibitor. In some embodiments, the hematological malignancy is DLBCL (Ramirez et al, "defines the causative factor that leads to activation of hedgehog signaling in diffuse large B-cell lymphoma)" leukemia study (leuk.res.) (2012), published online on 7 months and 17 days, and incorporated herein by reference in its entirety).
In some embodiments, the present disclosure provides a method of treating diffuse large B-cell lymphoma (DLBCL) comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: rituximab
Figure BDA0003526174380001981
Cyclophosphamide
Figure BDA0003526174380001982
Adriamycin
Figure BDA0003526174380001983
Vincristine
Figure BDA0003526174380001984
Prednisone, hedgehog signaling inhibitors, and combinations thereof.
In some embodiments, the present disclosure provides a method of treating multiple myeloma comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from: bortezomib
Figure BDA0003526174380001985
And dexamethasone
Figure BDA0003526174380001986
Hedgehog signaling inhibitors, BTK inhibitors, JAK/pan-JAK inhibitors, TYK2 inhibitors, PI3K inhibitors, SYK inhibitors and lenalidomide
Figure BDA0003526174380001987
Combinations of (a) and (b).
In some embodiments, the present disclosure provides a method of treating waldenstrom's macroglobulinemia: (
Figure BDA00035261743800019818
macrogolulinemia) comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: chlorambucil
Figure BDA0003526174380001988
Cyclophosphamide
Figure BDA0003526174380001989
Fludarabine (fludarabine)
Figure BDA00035261743800019810
Cladribine (cladribine)
Figure BDA00035261743800019811
Rituximab
Figure BDA00035261743800019812
Hedgehog signaling inhibitors, BTK inhibitors, JAK/pan-JAK inhibitors, TYK2 inhibitors, PI3K inhibitors, and SYK inhibitors.
In some embodiments, the present disclosure provides a method of treating alzheimer's disease comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: donepezil (donepezil)
Figure BDA00035261743800019813
Rivastigmine (rivastigmine)
Figure BDA00035261743800019814
Galantamine (galantamine)
Figure BDA00035261743800019815
Tacrine (tacrine)
Figure BDA00035261743800019816
Hemeijin (memantine)
Figure BDA00035261743800019817
In some embodiments, the present disclosure provides a method of treating organ transplant rejection or graft-versus-host disease comprising administering to a patient in need thereof a compound of formula I, II or III and one or more additional therapeutic agents selected from the group consisting of: steroids, cyclosporine, FK506, rapamycin, hedgehog signaling inhibitors, BTK inhibitors, JAK/pan-JAK inhibitors, TYK2 inhibitors, PI3K inhibitors, and SYK inhibitors.
In some embodiments, the disclosure provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I, II or III and a BTK inhibitor, wherein the disease is selected from inflammatory bowel disease, arthritis, Systemic Lupus Erythematosus (SLE), vasculitis, Idiopathic Thrombocytopenic Purpura (ITP), rheumatoid arthritis, psoriatic arthritis, osteoarthritis, Still's disease, juvenile arthritis, diabetes, myasthenia gravis, Hashimoto's thyroiditis, alder's thyroiditis, Graves ' disease, immunoautologous thyroiditis, sjogren's syndrome, multiple sclerosis, systemic sclerosis, neurolyme disease (lyurocortis, Guillain-barre syndrome), Guillain-barre syndrome (Guillain-barre myelitis), acute disseminated encephalomyelitis, or a BTK inhibitor, Edison's disease, ocular clonus-myoclonus syndrome, ankylosing spondylitis, antiphospholipid antibody syndrome, aplastic anemia, autoimmune hepatitis, autoimmune gastritis, pernicious anemia, celiac disease, Goodpasture's syndrome, idiopathic thrombocytopenic purpura, optic neuritis, scleroderma, primary biliary cirrhosis, Reiter's syndrome, Takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, Wegener's granulomatosis, psoriasis, alopecia universalis, Behcet's disease, chronic fatigue, autonomic imbalance, membranous glomerulonephropathy, endometriosis, interstitial cystitis, pemphigus vulgaris, bullous pemphigoid, greater neuromuscular mycosis, scleroderma, and ocular clonus syndrome, Vulvodynia, hyperproliferative diseases, rejection of transplanted organs or tissues, acquired immunodeficiency syndrome (AIDS, also known as HIV), type 1 diabetes, graft-versus-host disease, transplantation, infusion, systemic anaphylaxis, allergy (e.g., to plant pollen, latex, drugs, food, insect poison, animal hair, animal dander, dust mite or cockroach allergy), type I hypersensitivity, allergic conjunctivitis, allergic rhinitis, and atopic dermatitis, asthma, appendicitis, atopic dermatitis, asthma, allergy, blepharitis, bronchiolitis, bronchitis, bursitis, cervicitis, cholangitis, cholecystitis, chronic graft rejection, colitis, conjunctivitis, Crohn's disease, cystitis, dacryadenitis, dermatitis, dermatomyositis, encephalitis, endocarditis, endometritis, Enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, allergic purpura, hepatitis, hidradenitis suppurativa, immunoglobulin A nephropathy, interstitial lung disease, laryngitis, mastitis, meningitis, myelitis myocarditis, myositis, nephritis, oophoritis, orchitis, osteomyelitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonia (pneumoconiis), pneumonia (pneumoconia), polymyositis, proctitis, prostatitis, pyelonephritis, rhinitis, eustachyositis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, ulcerative colitis, uveitis, vaginitis, vasculitis or vulvitis, B cell proliferative disorders, such as diffuse large B cell lymphoma, follicular lymphoma, chronic lymphocytic leukemia, Acute lymphocytic leukemia, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma/Waldenstrom's macroglobulinemia, splenic marginal zone lymphoma, multiple myeloma (also known as plasma cell myeloma), non-Hodgkin's lymphoma, plasmacytoma, extranodal marginal zone B cell lymphoma, intranodal marginal zone B cell lymphoma, mantle cell lymphoma, mediastinal (thymic) large B cell lymphoma, intravascular large B cell lymphoma, primary effusion lymphoma, Burkitt's lymphoma/leukemia or lymphomatoid granulomatosis, breast cancer, prostate cancer or mast cell cancer (e.g., mast cell tumor, mast cell leukemia, mast cell sarcoma, systemic mastocytosis), bone cancer, colorectal cancer, pancreatic cancer, bone and joint diseases, including, but not limited to, rheumatoid arthritis, seronegative spondyloarthropathies (including ankylosing spondylitis, psoriatic arthritis, and Reiter's disease), behcet's disease, sjogren's syndrome, systemic sclerosis, osteoporosis, bone cancer metastasis, thromboembolic disorders, (e.g., myocardial infarction, angina, reocclusion after angioplasty, restenosis after angioplasty, reocclusion after aortic coronary bypass, restenosis after aortic coronary bypass, stroke, transient ischemia, peripheral arterial occlusion, pulmonary embolism, deep vein thrombosis), inflammatory pelvic conditions, urethritis, skin sunburn, sinusitis, pneumonia, encephalitis, meningitis, myocarditis, nephritis, osteomyelitis, myositis, hepatitis, gastritis, enteritis, dermatitis, gingivitis, appendicitis, pancreatitis, Cholecystitis, agammaglobulinemia, psoriasis, allergy, Crohn's disease, irritable bowel syndrome, ulcerative colitis, Huggelian's disease, tissue transplant rejection, hyperacute rejection of transplanted organs, asthma, allergic rhinitis, Chronic Obstructive Pulmonary Disease (COPD), autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome), autoimmune alopecia, pernicious anemia, glomerulonephritis, dermatomyositis, multiple sclerosis, scleroderma, vasculitis, autoimmune hemolytic and thrombocytopenic conditions, Goodpasture's syndrome, atherosclerosis, Edison's disease, Parkinson's disease, Alzheimer's disease, diabetes, septic shock, Systemic Lupus Erythematosus (SLE), rheumatoid arthritis, psoriatic arthritis, juvenile arthritis, osteoarthritis, chronic idiopathic thrombocytopenic purpura, Alzheimer's disease, Crohn's disease, inflammatory bowel disease, inflammatory disease, chronic idiopathic thrombocytopenic purpura, Crohn's disease, inflammatory bowel disease, inflammatory, Waldenstrom's macroglobulinemia, myasthenia gravis, hashimoto's thyroiditis, atopic dermatitis, degenerative joint disease, vitiligo, autoimmune hypopituitarism, guillain-barre syndrome, behcet's disease, scleroderma, mycosis fungoides, acute inflammatory responses (e.g., acute respiratory distress syndrome and ischemic/reperfusion injury), and graves' disease.
In some embodiments, the present disclosure provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I, II or III and a PI3K inhibitor, wherein the disease is selected from the group consisting of cancer, a neurodegenerative disorder, an angiogenic disorder, a viral disease, an autoimmune disease, an inflammatory disorder, a hormone-related disease, a condition associated with organ transplantation, an immunodeficiency disorder, a destructive bone disease, a proliferative disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, Chronic Myelogenous Leukemia (CML), Chronic Lymphocytic Leukemia (CLL), a liver disease, a pathological immune condition including T cell activation, a cardiovascular disorder, and a CNS disorder.
In some embodiments, the present disclosure provides a method of treating or lessening the severity of a disease comprising administering to a patient in need thereof a compound of formula I, II or III and a PI3K inhibitor, wherein the disease is selected from a benign or malignant tumor, a carcinoma or solid tumor of the brain, kidney (e.g., Renal Cell Carcinoma (RCC)), liver, adrenal gland, bladder, breast, stomach, ovary, colon, rectum, prostate, pancreas, lung, vagina, endometrium, cervix, testis, genitourinary tract, esophagus, throat, skin, bone, or thyroid, a sarcoma, glioblastoma, neuroblastoma, multiple myeloma, or gastrointestinal cancer, particularly a colon cancer or colorectal adenoma, or a tumor of the neck and head, epidermal hyperproliferation, psoriasis, prostatic hyperplasia, neoplasia, epithelial neoplasia, adenoma, a, Adenocarcinoma, keratoacanthoma, epidermoid tumor, large cell carcinoma, non-small cell lung carcinoma, lymphoma, (including, for example, non-hodgkin's lymphoma (NHL) and hodgkin's lymphoma (also known as hodgkin's disease or hodgkin's disease)), breast cancer, follicular cancer, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, or leukemia, diseases including Cowden syndrome (Cowden syndrome), lehmeter-dukes disease (lhemitte-Dudos disease), and Bannayan-zonner syndrome (Bannayan-Zonana syndrome), or diseases in which the PI3K/PKB pathway is abnormally activated; asthma of any type or origin, including intrinsic (non-allergic) asthma and extrinsic (allergic) asthma, mild asthma, moderate asthma, severe asthma, bronchitic asthma, exercise-induced asthma, occupational asthma and asthma induced following bacterial infection; acute Lung Injury (ALI), adult/Acute Respiratory Distress Syndrome (ARDS), chronic obstructive pulmonary, respiratory or pulmonary disease (COPD, COAD or COLD), including chronic bronchitis or asthma, emphysema associated therewith; and exacerbation of airway hyperreactivity following other drug therapies, particularly other inhaled drug therapies; bronchitis of any type or origin, including, but not limited to, acute, arachis, catarrhal (catarrhal), croupus (croupus), chronic or tuberculous bronchitis; pneumoconiosis (inflammatory, commonly occupational, lung disease, often accompanied by respiratory obstruction (whether chronic or acute), and caused by repeated inhalation of dust) of any type or origin, including, for example, aluminosis, charcoal-dust disease, asbestosis, lithosis, eyelash exfoliation, ferrosis, silicosis, tobacco-dust disease, and cotton-dust disease; loffler's syndrome; eosinophilia, pneumonia, parasitic (especially multicellular animal) infections (including tropical eosinophilia); bronchopulmonary aspergillosis; polyarteritis nodosa (including Churg-Strauss syndrome)); eosinophilic granuloma and eosinophil-related disorders affecting the respiratory tract resulting from drug response; psoriasis; contact dermatitis; atopic dermatitis; alopecia areata; erythema multiforme; dermatitis herpetiformis; scleroderma; leukoderma; hypersensitivity vasculitis; urticaria; bullous pemphigoid; lupus erythematosus; pemphigus; acquired epidermolysis bullosa; conjunctivitis; dry eye syndrome; and vernal conjunctivitis; diseases affecting the nose, including allergic rhinitis; and inflammatory diseases in which autoimmune reactions are involved or have an autoimmune component or etiology, including autoimmune blood disorders (e.g., hemolytic anemia, aplastic anemia, pure red cell anemia, and idiopathic thrombocytopenia); systemic lupus erythematosus; rheumatoid arthritis; polychondritis; scleroderma; wegener's granuloma; dermatomyositis; chronic active hepatitis; myasthenia gravis; Stefin-Johnson syndrome (Steven-Johnson syndrome); idiopathic sprue; autoimmune inflammatory bowel disease (e.g., ulcerative colitis and crohn's disease); endocrine ocular disorders; gray's disease; sarcoidosis; alveolitis; chronic allergic pneumonia; multiple sclerosis; primary biliary cirrhosis, uveitis (anterior and posterior), dry eye and vernal keratoconjunctivitis, interstitial pulmonary fibrosis, psoriatic arthritis and glomerulonephritis (with or without nephrotic syndrome, including, for example, idiopathic nephrotic syndrome or minor changes in nephropathy, restenosis, cardiac hypertrophy, atherosclerosis, myocardial infarction, ischemic stroke and congestive heart failure; alzheimer's disease; parkinson's disease; amyotrophic lateral sclerosis; Huntington's disease; and cerebral ischemia; and neurodegenerative diseases caused by traumatic injury, glutamate neurotoxicity or hypoxia.
The compounds and compositions of the present disclosure can be administered using any amount and any route of administration effective to treat or reduce the severity of cancer or a proliferative disorder. The exact amount required may vary from subject to subject, depending on the species, age and general condition of the subject, severity, particular agent, mode of administration, and the like. In some embodiments, the compounds are formulated in unit dosage forms that are easy to administer and are uniform in dosage. In some embodiments, a unit dosage form refers to a physically discrete unit of medicament suitable for the patient to be treated. It will be understood, however, that the total daily amount of the compounds and compositions will be decided by the attending physician within the scope of sound medical judgment. The specific effective dosage level for any particular subject or organism may depend upon a variety of factors, including the condition, disorder and disease being treated and its severity; the activity and/or characteristics of the particular compound used; the particular composition used; the age, weight, general health, sex, and diet of the subject; time of administration, route of administration, and/or rate of excretion of the particular compound used; the duration of treatment; drugs used in combination or concomitantly with the specific compound employed; and similar factors well known in the medical arts. In some embodiments, the patient is an animal, preferably a mammal, and most preferably a human.
Pharmaceutically acceptable compositions may be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (e.g., by powders, ointments, or drops), buccally, as an oral or nasal spray, and the like, depending on the severity of the infection being treated. In certain embodiments, the compounds may be administered orally or parenterally at dosage levels of about 0.01mg to about 50mg, and in some embodiments, about 1mg to about 25mg, per kilogram of subject body weight per day, one or more times per day, to achieve the desired therapeutic effect.
Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. In addition to inert diluents, the oral compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Injectable preparations, for example sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1, 3-butanediol. Acceptable vehicles and solvents that can be employed are water, ringer's solution, U.S. p. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono-or diglycerides. In addition, fatty acids, such as oleic acid, are used in the preparation of injectables.
The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter or by incorporation of sterilizing agents, in the form of sterile solid compositions that can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
In order to prolong the effect of the compounds of the present disclosure, it is often desirable to slow the absorption of the subcutaneously or intramuscularly injected compounds. This can be achieved by using liquid suspensions of crystalline or amorphous materials with poor water solubility. The rate of absorption of the compound then depends on its rate of dissolution, which in turn may depend on the crystal size and crystalline form. Alternatively, delayed absorption of the parenterally administered compound form is achieved by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are prepared by forming a microcapsule matrix of the compound in a biodegradable polymer, such as polylactide-polyglycolide. Depending on the ratio of compound to polymer and the nature of the particular polymer employed, the release rate of the compound can be controlled. Examples of other biodegradable polymers include poly (orthoesters) and poly (anhydrides). Depot injectable formulations are also prepared by entrapping the compounds in liposomes or microemulsions which are compatible with body tissues.
In some embodiments, compositions for rectal or vaginal administration are suppositories which can be prepared by mixing the compound with a suitable non-irritating excipient or carrier, for example cocoa butter, polyethylene glycol or a suppository wax, which is solid at ambient temperature but liquid at body temperature and therefore melts in the rectum or vaginal cavity and releases the active compound.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In such solid dosage forms, the active compound is admixed with: at least one inert pharmaceutically acceptable excipient or carrier, such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and silicic acid; b) binders such as carboxymethyl cellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and acacia; c) humectants, such as glycerol; d) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates and sodium carbonate; e) solution retarders, such as paraffin; f) absorption accelerators, such as quaternary ammonium compounds; g) wetting agents, such as cetyl alcohol and glycerol monostearate; h) absorbents such as kaolin and bentonite clay; and i) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate and mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid compositions of a similar type may also be employed as fillers in soft-filled gelatin capsules and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. Tablets, dragees, capsules, pills and granules of solid dosage forms can be prepared with coatings and shells such as enteric coatings and other coatings well known in the art of pharmaceutical formulation. It may optionally contain opacifying agents and may also have a composition such that it releases only or preferentially one or more active ingredients, optionally in a certain portion of the intestinal tract, in a delayed manner. Examples of embedding compositions that may be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft-filled gelatin capsules and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
The active compound may also be in microencapsulated form with one or more excipients as mentioned above. Tablets, dragees, capsules, pills and granules of solid dosage forms can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the art of pharmaceutical formulation. In such solid dosage forms, the active compound may be mixed with at least one inert diluent (e.g., sucrose, lactose or starch). Such dosage forms may also include, as is normal practice, additional substances other than inert diluents, such as tableting lubricants and other tableting aids, for example magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. It may optionally contain opacifying agents and may also have a composition such that it releases only or preferentially one or more active ingredients, optionally in a certain portion of the intestinal tract, in a delayed manner. Examples of embedding compositions that may be used include polymeric substances and waxes.
Dosage forms for topical or transdermal administration of the compounds include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier and any required preservatives or buffers as required. In some embodiments, the composition is an ophthalmic formulation, ear drops, or eye drops. In addition, the present disclosure encompasses the use of transdermal patches, which have the added advantage of allowing controlled delivery of the compound to the body. The dosage form may be manufactured by dissolving or dispensing the compound in a suitable medium. Absorption enhancers may also be used to increase the flux of the compound across the skin. The rate can be controlled by providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.
In some embodiments, the present disclosure provides a method of inhibiting protein kinase activity in a biological sample comprising the step of contacting the biological sample with a compound of the present disclosure or a composition comprising the compound.
In some embodiments, the biological sample comprises, but is not limited to, a cell culture or extract thereof; a biopsy material obtained from a mammal or an extract thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
Depending on the particular condition or disease to be treated, additional therapeutic agents typically administered to treat the condition may also be present in the compositions of the present disclosure. As used herein, an additional therapeutic agent normally administered for the treatment of a particular disease or condition is referred to as "appropriate for the disease or condition being treated.
Furthermore, the compounds and/or compositions of the present disclosure may be used in combination therapy, i.e., the compounds and/or compositions of the present disclosure may be administered simultaneously with, prior to, or after one or more other therapeutic agents or medical procedures, particularly for the treatment of various cancers. In some embodiments, the compounds of the present disclosure may also be advantageously used in combination with other anti-proliferative compounds. The particular combination of therapies (therapeutic agents or procedures) used in a combination regimen will take into account the compatibility of the other therapeutic agents and/or procedures required and the desired therapeutic effect to be achieved. It will also be appreciated that the therapies used may achieve the desired effect against the same condition (e.g., the compound provided may be administered simultaneously with another anti-cancer agent), or that the therapies may achieve different effects (e.g., control of any adverse effects). In some embodiments, the therapeutic agent is a chemotherapeutic agent or an anti-proliferative compound. Exemplary chemotherapeutic agents include, but are not limited to, alkylating agents, nitrosourea agents, antimetabolites, antitumor antibiotics, plant-derived alkaloids, topoisomerase inhibitors, hormonal therapy drugs, hormone antagonists, aromatase inhibitors, P-glycoprotein inhibitors, platinum complex derivatives, other immunotherapeutic drugs, and other anticancer agents. In addition, the provided technology can be used with or prepared as a mixture with leukopenia (neutrophil) drugs, thrombocytopenia drugs, antiemetic drugs, and cancer pain drugs for QOL recovery in patients as cancer treatment adjuvants. In some embodiments, the therapeutic agent is an antibody. In some embodiments, the therapeutic agent is an immunomodulatory agent. In some embodiments, the immunomodulator targets a cell surface signaling molecule on an immune cell. In some embodiments, the immunomodulatory agent targets a cell surface signaling molecule on an immune cell, wherein the agent is an antagonist that blocks a co-inhibitory pathway. In some embodiments, the immune modulator is a checkpoint blockade agent. In some embodiments, the immunomodulator is an antibody that targets a cell surface signaling protein expressed by an immune cell. In some embodiments, the immunomodulatory agent is an antibody that targets a protein selected from PD-1, PD-L1, CTLA4, TIGIT, BTLA, TIM-3, LAG3, B7-H3, and B7-H4. In some embodiments, the immunomodulator is a PD-1 antibody (e.g., nivolumab, pembrolizumab, pidilizumab, BMS 936559, MPDL328OA, etc.). In some embodiments, the immunomodulator is a PD-L1 antibody. In some embodiments, the immunomodulatory agent is a CTLA4 antibody (e.g., ipilimumab). In some embodiments, the immunomodulatory agent is a TIGIT antibody. In some embodiments, the immunomodulatory agent is a BTLA antibody. In some embodiments, the immunomodulator is a Tim-3 antibody. In some embodiments, the immunomodulatory agent is a LAG3 antibody. In some embodiments, the immunomodulator is a B7-H3 antibody. In some embodiments, the immunomodulator is a B7-H4 antibody. In some embodiments, the immunomodulator targets a cell surface signaling molecule on an immune cell, wherein the agent is an agonist involved in a co-stimulatory pathway. In some embodiments, such immunomodulatory agents are or include antibodies that target co-stimulatory receptors. In some embodiments, the antibody activates a T cell co-stimulatory receptor. In some embodiments, the antibody targets a member of the Tumor Necrosis Factor (TNF) receptor superfamily. In some embodiments, the antibody targets a protein selected from CD137(4-1BB), CD357(GITR, TNFRS18, AITR), CD134(OX40), and CD40 (TNFRSF 5). In some embodiments, the antibody is an anti-CD 137 antibody (e.g., ureluzumab). In some embodiments, the antibody is an anti-CD 357 antibody. In some embodiments, the antibody is an anti-CD 40 antibody. In some embodiments, the antibody is an anti-CD 134 antibody. Additional exemplary T cell co-stimulatory and co-inhibitory receptors are described in Chen L, Flies DB., the Molecular mechanisms of T cell co-stimulation and co-inhibition (Molecular mechanisms of T cell co-stimulation and co-inhibition), nature review immunology (nat. rev. immunol.)2013, 13(4), 227-42 and Yao S et al, Advances in targeting cell surface signaling molecules for immunomodulation (advancement in targeting cell surface signaling molecules for immunization modification), nature review drug discovery (nat. rev. drug discovery) 2013, 12(2), 136-40. In some embodiments, the therapeutic agent is an antibody that activates such stimulatory receptors or blocks such inhibitory receptors.
In some embodiments, the one or more additional therapeutic agents are or include tumor-specific immune cells. In some embodiments, the one or more additional therapeutic agents are or include tumor-specific T cells. In some embodiments, the one or more additional therapeutic agents is or includes Tumor Infiltrating Lymphocytes (TILs). In some embodiments, the one or more additional therapeutic agents are or comprise T cells ectopically expressing a known anti-tumor T Cell Receptor (TCR). In some embodiments, the one or more additional therapeutic agents is or includes a Chimeric Antigen Receptor (CAR) T cell. In some embodiments, provided compositions include an immune enhancing substance. Exemplary immune enhancing substances that can be used in combination with the provided compounds, compositions, and/or methods include, but are not limited to, various cytokines and tumor antigens. Cytokines that stimulate an immune response include, for example, GM-CSF, M-CSF, G-CSF, interferon- α, interferon- β, interferon- γ, IL-1, IL-2, IL-3, and IL-12, and the like. Antibodies that block inhibitory receptors and/or activate stimulatory receptors, such as, but not limited to, B7 ligand derivatives, anti-CD 3 antibodies, anti-CD 28 antibodies, and anti-CTLA-4 antibodies, are also known in the art and described herein, can improve immune responses. In some embodiments, the therapeutic agent is a small molecule for immunomodulation. In some embodiments, the therapeutic agent is a small molecule that mediates antitumor immune activity. In some embodiments, the therapeutic agent is a small molecule that targets an enzyme directly involved in immune modulation. In some embodiments, the therapeutic agent is an indoleamine 2, 3-dioxygenase (IDO) inhibitor. In some embodiments, the therapeutic agent is an IDO1 inhibitor, such as F001287, indoimod (indoximod), NLG-919, and INCB 024360. In some embodiments, the therapeutic agent is a tryptophan-2, 3 dioxygenase (TDO) inhibitor. In some embodiments, the therapeutic agent is a dual IDO/TDO inhibitor. In some embodiments, the therapeutic agent is an IDO selective inhibitor. In some embodiments, in some other embodiments, the therapeutic agent is a TDO selective inhibitor. In some embodiments, provided compositions include an IDO inhibitor and a first construct. In some embodiments, provided compositions include an IDO inhibitor, a first construct, and a second construct. It will be appreciated that the immune response to the first construct and/or the second construct may be significantly enhanced by administration of an IDO inhibitor.
In some embodiments, medical procedures that can be used in combination with the compounds, compositions, and methods of the present application include, but are not limited to, surgery, radiotherapy (gamma-irradiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioisotopes, to name a few), endocrine therapy, biological response modifiers (interferons, interleukins, and Tumor Necrosis Factor (TNF), to name a few), hyperthermia, cryotherapy, and adoptive T cell transfer (e.g., TIL therapy, transgenic TCR, CAR T cell therapy, NK cell therapy, etc.). In some embodiments, the medical procedure is surgery. In some embodiments, the medical procedure is radiotherapy.
In some embodiments, the provided techniques include provided agents (e.g., provided agents that include moieties that can bind to CD 38) and cell populations (e.g., cells of a cell-based therapy). In some embodiments, provided techniques include provided agents and effector cell populations. In some embodiments, the cell population is a manipulated cell population, e.g., that is typically engineered, activated, enriched, and/or expanded, etc. In some embodiments, the cell population is a manufactured cell population. In some embodiments, the cell population is enriched for certain types of desired cells. In some embodiments, the effector cell is an NKT cell. In some embodiments, the effector cell is a monocyte. In some embodiments, the effector cell is a macrophage. In some embodiments, the effector cell is a T cell. In some embodiments, the effector cell is a CAR T cell. In some embodiments, the effector cell is an NK cell, e.g., a cytokine-induced memory-like NK cell, a CAR-NK cell, an engineered NK cell that expresses or does not express certain proteins (e.g., receptors), and can be from a variety of sources. As will be appreciated by those skilled in the art, suitable immune cells (e.g., NK cells) can be derived from a variety of sources and/or engineered in a variety of ways. For example, in some embodiments, the NK cells are memory-like NK cells. In some embodiments, the NK cells are cytokine-induced memory-like NK cells. In some embodiments, the NK cells are derived from stem cells. In some embodiments, the NK cells are derived from an iPSC cell line. In some embodiments, the NK cells are derived from a clonal master iPSC cell line. In some embodiments, the NK cells are engineered to express certain receptors, such as the high affinity, optionally non-cleavable, CD16 receptor. In some embodiments, the NK cell is engineered to express a Chimeric Antigen Receptor (CAR), e.g., in some embodiments, the NK cell can be engineered to express an anti-CD 19 CAR. In some embodiments, the NK cell is a CAR-NK cell. In some embodiments, the NK cell is engineered to express a cytokine receptor. In some embodiments, the NK cells comprise IL-15 receptorbody that enhances persistence and expansion without the need for co-administration of cytokine loads. In some embodiments, the NK cells are engineered to prevent expression of certain cellular proteins (e.g., certain cell surface proteins). In some embodiments, the NK cell is engineered to prevent expression of CD 38. In some embodiments, the NK cells are derived from placenta. In some embodiments, the NK cell is a donor NK cell. In some embodiments, the NK cells are haploid concordant donor NK cells. In some embodiments, the NK cells are mismatched donor NK cells. In some embodiments, the NK cell is a related donor NK cell, e.g., a mismatched related donor NK cell. In some embodiments, the NK cell is an unrelated donor NK cell. In some embodiments, the NK cells are derived from a subject, e.g., a patient. In some embodiments, the provided techniques include a innate cell engager, such as one that binds to innate cells (e.g., NK cells and macrophages) while binding to specific tumor cells. In some embodiments, the NK cells are derived from cord blood stem cells and progenitor cells. In some embodiments, the NK cell is derived from the modulation of a signaling pathway (e.g., Notch signaling pathway). In some embodiments, the nanoparticles are utilized to improve and/or maintain the growth of NK cells. In some embodiments, as described herein, the NK cells are produced ex vivo. In some embodiments, NK cells can be cryopreserved and stored as a ready-made cell therapy in multiple doses. Examples of some such technologies include those utilized by fat Therapeutics, natkwest, cellularity, green cross pharmaceutical (GC Pharma), sorento Therapeutics, Inc, affimted/MD Anderson Cancer Center (MD Anderson Cancer Center), gamda Cell co. Those skilled in the art will appreciate that where such techniques can optionally be utilized, antibodies and/or CARs to particular antigens utilized in certain such techniques may not be required in provided techniques including ARM as described herein. In some embodiments, the provided agent, cell (e.g., effector cell), and optionally immunoglobulin (e.g., IgG (e.g., intravenous immunoglobulin)) are administered to a subject such that the subject can be exposed to the provided agent and cell. In some embodiments, the provided agent and the cell (e.g., effector cell) are administered simultaneously, either in a single composition (e.g., a composition comprising the provided agent, the cell (e.g., effector cell), and optionally the immunoglobulin), or separately; in some embodiments, the provided agents are administered before or after the cells (e.g., effector cells). In some embodiments, the present disclosure provides methods for treating various conditions, disorders or diseases (e.g., various cancers) comprising administering the provided agents and cells to a subject suffering from or susceptible to the condition, disorder or disease. In some embodiments, the cells (e.g., effector cells) are stored (e.g., cryopreserved) with the provided agent (e.g., ARM agent). In some embodiments, it is thawed and administered as a single composition. In some embodiments, one or more doses of cells (e.g., effector cells) are administered followed by a dose of the provided agent (e.g., an ARM agent). In some embodiments, the dose of agent provided is a single systemic dose. In some embodiments, multiple doses of cells (e.g., effector cells) are administered, followed by a dose (in some cases a single systemic dose) of the provided agent (e.g., an ARM agent). In some embodiments, an agent (e.g., an ARM agent) is administered (e.g., systemically, in some cases a single systemic administration), followed by one or more (in some cases multiple) doses of cells (e.g., effector cells). In some embodiments, an agent (e.g., an ARM agent) is administered (e.g., systemically, in some cases a single systemic administration), followed by one or more (in some cases multiple) each independent dose of cells (e.g., effector cells) or cells and provided agent (e.g., ARM agent), wherein the cells and provided agent can be administered in a combined composition or in separate compositions.
Further, such provided techniques may provide enhanced efficacy and/or reduced side effects.
Anti-proliferative compounds include, but are not limited to, aromatase inhibitionAn agent; an antiestrogen; a topoisomerase I inhibitor; a topoisomerase II inhibitor; a microtubule active compound; an alkylating compound; (ii) a histone deacetylase inhibitor; compounds that induce a cellular differentiation process; a cyclooxygenase inhibitor; an MMP inhibitor; an mTOR inhibitor; antineoplastic antimetabolites; a platinum compound; compounds that target/reduce protein or lipid kinase activity and other anti-angiogenic compounds; a compound that targets, reduces or inhibits the activity of a protein or lipid phosphatase; a gonadorelin (gonadorelin) agonist; an antiandrogen; a methionine aminopeptidase inhibitor; a matrix metalloproteinase inhibitor; a bisphosphonate; a biological response modifier; an anti-proliferative antibody; a heparinase inhibitor; inhibitors of Ras oncogenic isoforms; a telomerase inhibitor; a proteasome inhibitor; compounds for use in the treatment of hematological malignancies; a compound that targets, decreases or inhibits the activity of Flt-3; hsp90 inhibitors, such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Comfortma Therapeutics; temozolomide (temozolomide)
Figure BDA0003526174380002081
Spindle kinesin inhibitors, such as SB715992 or SB743921 from glatiramer (GlaxoSmithKline), or pentamidine (pentamidine)/chlorpromazine from portentos (CombinatoRx); MEK inhibitors, such as ARRY142886 from alay biopharmaceutical (Array BioPharma), AZD6244 from astrikon (AstraZeneca), PD181461 from Pfizer, and leucovorin. As used herein, the term "aromatase inhibitor" relates to a compound that inhibits estrogen production, e.g. the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively. The term includes, but is not limited to steroids, especially atamestane (atamestane), exemestane (exemestane), and formestane (formestane); and in particular non-steroids, in particular aminoglutethimide, roglucimide, pirglutethimide, troloxacillinTandane, testolactone, ketoconazole, vorozole, fadrozole, anastrozole and letrozole. Exemestane is available under the trade name AromasinTMAnd (5) selling. Formestane is under the trade name Lentaron TMAnd (5) selling. Fadrozole is given the trade name AfemaTMAnd (5) selling. Anastrozole is given the trade name ArimidexTMAnd (5) selling. Letrozole is given the trade name FemaraTMOr FemarTMAnd (5) selling. Aminoglutethimide under the trade name OrimetenTMAnd (5) selling. Combinations of the present disclosure that include chemotherapeutic agents that are aromatase inhibitors are particularly useful for treating hormone receptor positive tumors, such as breast tumors.
In some embodiments, the antiestrogen is a compound that antagonizes the effects of estrogen at the estrogen receptor level. The term includes, but is not limited to, tamoxifen (tamoxifen), fulvestrant (fulvestrant), raloxifene (raloxifene), and raloxifene hydrochloride. Tamoxifen is available under the trade name NolvadexTMAnd (5) selling. Raynaxiphenol hydrochloride is under the trade name EvistaTMAnd (5) selling. Fulvestrant may be under the brand name FaslodexTMAnd (4) application. Combinations of the present disclosure that include chemotherapeutic agents that are anti-estrogens are particularly useful for treating estrogen receptor positive tumors, such as breast tumors.
In some embodiments, the antiandrogen is a substance capable of inhibiting the biological effects of androgens and includes, but is not limited to, bicalutamide (Casodex)TM). As used herein, the term "gonadoliberin agonist" includes, but is not limited to abarelix (abarelix), goserelin (goserelin), and goserelin acetate. Goserelin may be under the trade name Zoladex TMAnd (4) application.
In some embodiments, topoisomerase I inhibitors include, but are not limited to, topotecan (topotecan), gimatecan (gimatecan), irinotecan (irinotecan), camptothecin (camptothecan), and its analog 9-nitrocamptothecin and macromolecular camptothecin conjugate PNU-166148. Irinotecan may be, for example, in its form of sale (e.g. under the trademark Camptosar)TM) And (4) application. Topotecan is known under the trade name HycamptinTMAnd (5) selling.
In some embodiments, the topoisomerase II inhibitor comprises, but is not limited to, an anthracycline, such as doxorubicin (comprising a liposome formulation, such as Caelyx)TM) Daunorubicin (daunorubicin), epirubicin (epirubicin), idarubicin (idarubicin) and nemorubicin (nemorubicin), anthraquinone mitoxantrone and losoxantrone (losoxantrone), and etoposide (etoposide) and teniposide (teniposide). Etopophos is the trade name EtopophosTMAnd (5) selling. Teniposide is sold under the trade name VM 26-Bristol. Adriamycin is known under the trade name Acribilastin TMOr AdriamycinTMAnd (5) selling. Epirubicin is known under the trade name FarmorubicinTMAnd (5) selling. Idarubicin is available under the trade name ZavedosTMAnd (5) selling. Mitoxantrone is sold under the trade name Novantron.
In some embodiments, the microtubule active agents are microtubule stabilizing, microtubule destabilizing compounds and tubulin (microtubulin) polymerization inhibitors, including but not limited to taxanes (taxanes), such as paclitaxel (paclitaxel) and docetaxel (docetaxel); vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine (vinorelbine); dieschorlide (discodermolide); colchicine and epothilone (epothilone) and derivatives thereof. Taxol is given the trade name TaxolTMAnd (5) selling. Docetaxel having the trade name TaxotereTMAnd (5) selling. Vinblastine sulfate is under the trade name Vinblasttin R.PTMAnd (5) selling. Vincristine sulfate is known under the trade name FarmistinTMAnd (5) selling.
In some embodiments, the alkylating agent comprises, but is not limited to, cyclophosphamide, ifosfamide, melphalan, or nitrosourea (BCNU or Gliadel). Cyclophosphamides are known under the trade name cyclosatinsTMAnd (5) selling. Ifosfamide is known under the trade name HoloxanTMAnd (5) selling.
In some embodiments, the histone deacetylase inhibitor or HDAC inhibitor is a compound that inhibits histone deacetylase and has antiproliferative activity. This includes, but is not limited to suberoylanilide hydroxamic acid (SAHA).
In some embodiments, the antineoplastic antimetabolite comprisesBut are not limited to 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds (e.g., 5-azacytidine and decitabine), methotrexate and edatrexate, and folic acid antagonists (e.g., pemetrexed). Capecitabine is under the trade name XelodaTMAnd (5) selling. Gemcitabine under the trade name GemzarTMAnd (5) selling.
In some embodiments, platinum compounds include, but are not limited to, carboplatin (carboplatin), cisplatin (cis-platinum), cisplatin (cissplatinum), and oxaliplatin (oxaliplatin). Carboplatin can be sold, for example, under the trademark CarboplatTMAnd (4) application. Oxaliplatin can be sold, for example, under the trademark EloxatinTMAnd (4) application.
In some embodiments, protein or lipid kinase activity is targeted/reduced; or a protein or lipid phosphatase activity; or other anti-angiogenic compounds include, but are not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds that target, decrease or inhibit the activity of Platelet Derived Growth Factor Receptor (PDGFR), such as compounds that target, decrease or inhibit the activity of PDGFR, especially compounds that inhibit PDGF receptor, such as N-phenyl-2-pyrimidine-amine derivatives, such as imatinib (imatinib), SU101, SU6668 and GFB-111; b) a compound that targets, reduces or inhibits the activity of a Fibroblast Growth Factor Receptor (FGFR); c) a compound that targets, reduces or inhibits the activity of insulin-like growth factor receptor I (IGF-IR), for example a compound that targets, reduces or inhibits the activity of IGF-IR, in particular a compound that inhibits the kinase activity of IGF-I receptor, or an antibody that targets the extracellular domain of IGF-I receptor or its growth factor; d) a compound that targets, reduces or inhibits the activity of the Trk receptor tyrosine kinase family, or an ephrin B4 inhibitor; e) a compound that targets, reduces or inhibits the activity of the AxI receptor tyrosine kinase family; f) a compound that targets, decreases or inhibits the activity of Ret receptor tyrosine kinase; g) compounds that target, decrease or inhibit the activity of Kit/SCFR receptor tyrosine kinases, such as imatinib; h) targeting, lowering Compounds that reduce or inhibit the activity of the C-Kit receptor tyrosine kinase that is part of the PDGFR family, such as compounds that target, reduce or inhibit the activity of the C-Kit receptor tyrosine kinase family, especially compounds that inhibit the C-Kit receptor, such as imatinib; i) compounds that target, decrease or inhibit the activity of c-Abl family members, their gene fusion products (e.g., BCR-Abl kinase) and mutants, e.g., compounds that target, decrease or inhibit the activity of c-Abl family members and their gene fusion products, e.g., N-phenyl-2-pyrimidine-amine derivatives, e.g., imatinib or nilotinib (AMN 107); PD 180970; AG 957; NSC 680410; PD173955 from parkievis (ParkeDavis); or dasatinib (BMS-354825); j) compounds that target, reduce or inhibit the activity of a member of the Raf family of protein kinases c (pkc) and serine/threonine kinases, MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, TYK2, BTK and TEC families, and/or a member of the cyclin dependent kinase family (CDK), including staurosporine derivatives, such as midostaurin; examples of other compounds include UCN-01, safrog (safingol), BAY 43-9006, Bryostatin (Bryostatin)1, piperacillin (Perifosine); imofosine (llmofosine); RO 318220 and RO 320432; GO 6976; lsis 3521; LY333531/LY 379196; an isoquinoline compound; FTI; PD184352 or QAN697(P13K inhibitor) or AT7519(CDK inhibitor); k) compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors, for example compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors, comprising imatinib mesylate (Gleevec) TM) Or tofukast (tyrphostin), such as tofukast A23/RG-50810; AG 99; tafosistin AG 213; tafosastine AG 1748; tafosastine AG 490; tafosastine B44; tafosistine B44(+) enantiomer; tafosistin AG 555; AG 494; tafosiltin AG 556, AG957 and adaphtin (adaphstin) (4- { [ (2, 5-dihydroxyphenyl) methyl]Amino } -benzoic acid adamantane esters; NSC 680410, adafostine); 1) epidermal growth factor family (EGFR) that targets, reduces or inhibits receptor tyrosine kinases1ErbB2, ErbB3, ErbB4 in homo-or hetero-polymer formDimeric form) and mutants thereof, e.g. compounds that target, decrease or inhibit the activity of the epidermal growth factor receptor family, in particular compounds that inhibit members of the EGF receptor tyrosine kinase family, e.g. EGF receptor, ErbB2, ErbB3 and ErbB4, or bind to EGF or EGF-related ligands, proteins or antibodies, CP358774, ZD 1839, ZM 105180; trastuzumab (trastuzumab) (Herceptin)TM) Cetuximab (Erbitux)TM) Iressa, Tarceva, OSI-774, Cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.11, E6.3 or E7.6.3, and 7H-pyrrolo- [2, 3-d ] ]A pyrimidine derivative; m) a compound that targets, reduces or inhibits the activity of the c-Met receptor, e.g., a compound that targets, reduces or inhibits the activity of c-Met, particularly a compound that inhibits the kinase activity of the c-Met receptor, or an antibody that targets the extracellular domain of c-Met or binds HGF; n) compounds that target, decrease or inhibit the kinase activity of one or more JAK family members (JAK1/JAK2/JAK3/TYK2 and/or pan-JAK), including but not limited to PRT-062070, SB-1578, baritinib (baritinib), paritinib (pactinib), molotetinib (momelotinib), VX-509, AZD-1480, TG-101348, tofacitinib, and lucolintinib; o) compounds that target, decrease or inhibit the kinase activity of PI3 kinase (PI3K), including but not limited to ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib (buparlisib), pitrielixib (pictelisib), PF-4691502, BYL-719, dalutoxib (dactylisib), XL-147, XL-765, and idecoxib (idelalisib); and q) compounds that target, decrease or inhibit the signaling effect or smoothing receptor (SMO) pathway of hedgehog (Hh), including but not limited to cyclopamine, vismodegib (vismodegib), itraconazole (itraconazole), imodegi (eriodegib), and IPI-926 (saridegib).
In some embodiments, PI3K inhibitors include, but are not limited to, compounds having inhibitory activity against one or more enzymes of the phosphatidylinositol-3-kinase family, including, but not limited to, PI3K α, PI3K γ, PI3K δ, PI3K β, PI3K-C2 α, PI3K-C2 β, PI3K-C2 γ, Vps34, p110- α, p110- β, p110- γ, p110- δ, p85- α, p85- β, p55- γ, p150, p101, and p 87. Examples of PI3K inhibitors include, but are not limited to, ATU-027, SF-1126, DS-7423, PBI-05204, GSK-2126458, ZSTK-474, buparxib, Pitrixib, PF-4691502, BYL-719, daluxib, XL-147, XL-765, and idexib.
In some embodiments, the BTK inhibitor comprises, but is not limited to, a compound having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including, but not limited to, AVL-292 and ibrutinib.
In some embodiments, SYK inhibitors include, but are not limited to, compounds having inhibitory activity against spleen tyrosine kinase (SYK), including, but not limited to, PRT-062070, R-343, R-333, Iselier (Excellair), PRT-062607, and fostatinib (fostamatinib).
Further examples of BTK inhibiting compounds and conditions that can be treated by the compounds in combination with the disclosed compounds can be found in WO2008039218 and WO2011090760, the entire contents of which are incorporated herein by reference.
Further examples of SYK inhibiting compounds and conditions that may be treated by the compounds in combination with the disclosed compounds may be found in WO2003063794, WO2005007623 and WO2006078846, the entire contents of which are incorporated herein by reference.
Further examples of PI3K inhibiting compounds and conditions that can be treated by the compounds in combination with the disclosed compounds can be found in WO2004019973, WO2004089925, WO2007016176, US8138347, WO2002088112, WO2007084786, WO2007129161, WO2006122806, WO2005113554 and WO2007044729, the entire contents of which are incorporated herein by reference.
Other examples of JAK-inhibiting compounds and conditions that can be treated by the compounds in combination with the presently disclosed compounds can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246, and WO2007070514, the entire contents of which are incorporated herein by reference.
Other anti-angiogenic compounds include those that have another mechanism of activity,for example compounds not related to protein or lipid kinase inhibition, such as thalidomide (Thalomid)TM) And TNP-470.
Examples of proteasome inhibitors suitable for use in combination with the compounds of the present disclosure include, but are not limited to, bortezomib, disulfiram (disulfiram), epigallocatechin-3-gallate (EGCG), salinosporin A, carfilzomib, ONX-0912, CEP-18770, and MLN 9708.
Compounds targeting, decreasing or inhibiting the activity of a protein or lipid phosphatase are for example inhibitors of phosphatase 1, phosphatase 2A or CDC25, such as okadaic acid (okadaic acid) or derivatives thereof.
Compounds that induce a cellular differentiation process include, but are not limited to, retinoic acid, alpha-, gamma-, or delta-tocopherol, or alpha-, gamma-, or delta-tocotrienol.
In some embodiments, the cyclooxygenase inhibitor comprises, but is not limited to, Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acids and derivatives, such as celecoxib (Celebrex)TM) Rofecoxib (Vioxx)TM) Etoricoxib, valdecoxib or 5-alkyl-2-arylaminophenylacetic acids, such as 5-methyl-2- (2 '-chloro-6' -fluoroanilino) phenylacetic acid, lumiracoxib.
In some embodiments, bisphosphonates include, but are not limited to, itraconazole (ethidonic acid), clodronic acid (clodronic acid), tiludronic acid (tiludronic acid), pamidronic acid (pamidronic acid), alendronic acid (alendronic acid), ibandronic acid (ibandronic acid), risedronic acid (risedronic acid), and zoledronic acid (zoledronic acid). Didronic acid under the trade name DidronelTMAnd (5) selling. Chlorophosphonic acids under the trade name Bonefos TMAnd (5) selling. Telophosphonic acid under the trade name SkelidTMAnd (5) selling. Pamidronic acid is under the trade name ArediaTMAnd (5) selling. Alendronic acid under the trade name FosamaxTMAnd (5) selling. Ibandronic acid is given the trade name BondranatTMAnd (5) selling. Risedronic acid under the trade name ActonelTMAnd (5) selling. Zomet phosphonic acid under the trade name ZometTMAnd (5) selling. In some embodiments, mTOR inhibitsThe agent is a compound that inhibits mammalian target of rapamycin (mTOR) and has antiproliferative activity, such as sirolimus
Figure BDA0003526174380002131
Everolimus (Certican)TM) CCI-779 and ABT 578.
In some embodiments, the heparinase inhibitor is a compound that targets, reduces or inhibits the degradation of heparin sulfate. Including but not limited to PI-88. In some embodiments, the biological response modifier is a lymphokine or an interferon.
In some embodiments, inhibitors of Ras oncogenic isoforms (e.g., H-Ras, K-Ras, or N-Ras) are compounds that target, decrease, or inhibit the oncogenic activity of Ras; for example "farnesyl transferase inhibitors", for example L-744832, DK8G557 or R115777 (Zarnestra)TM). In some embodiments, the telomerase inhibitor is a compound that targets, decreases, or inhibits telomerase activity. Compounds which target, decrease or inhibit telomerase activity are in particular compounds which inhibit the telomerase receptor, for example telomerase (telomestatin).
In some embodiments, the methionine aminopeptidase inhibitor is a compound that targets, reduces, or inhibits methionine aminopeptidase activity. Compounds that target, decrease or inhibit methionine aminopeptidase activity include, but are not limited to, benguanamide (bengamide) or derivatives thereof.
In some embodiments, the proteasome inhibitor is a compound that targets, decreases, or inhibits proteasome activity. Compounds that target, decrease or inhibit proteasome activity include, but are not limited to, bortezomib (Velcade)TM) And MLN 341.
In some embodiments, matrix metalloproteinase inhibitors or ("MMP" inhibitors) include, but are not limited to, collagen peptide mimetic and non-peptidomimetic inhibitors, tetracycline derivatives, such as the oxamido peptidomimetic inhibitor batimastat (batimastat) and its analogs with oral bioavailability marimastat (marimastat) (BB-2516), prinomastat (prinomastat) (AG3340), metamastat (metastat) (NSC 683551), BMS-279251, BAY 12-9566, TAA211, MMI270B, or AAJ 996.
In some embodiments, the compound for treating hematological malignancies comprises, but is not limited to, an FMS-like tyrosine kinase inhibitor, which is a compound that targets, decreases or inhibits the activity of the FMS-like tyrosine kinase receptor (Flt-3R); interferon, 1-beta-D-arabinofuranosyl cytosine (ara-c) and busulfan; and ALK inhibitors, which are compounds that target, decrease or inhibit anaplastic lymphoma kinase.
Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, staurosporine derivatives, SU11248 and MLN 518.
In some embodiments, HSP90 inhibitors include, but are not limited to, compounds that target, decrease or inhibit the intrinsic ATPase (ATPase) activity of HSP 90; compounds that degrade, target, reduce or inhibit HSP90 client proteins (client proteins) via the ubiquitin proteasome pathway. Compounds which target, reduce or inhibit the intrinsic atpase activity of HSP90 are in particular compounds, proteins or antibodies which inhibit the atpase activity of HSP90, such as 17-allylamino, 17-dimethoxy geldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol (radicicol); and HDAC inhibitors.
In some embodiments, the anti-proliferative antibody comprises, but is not limited to, trastuzumab (Herceptin)TM) trastuzumab-DM 1, erbitux, bevacizumab AvastinTM) Rituximab, and methods of use
Figure BDA0003526174380002141
PRO64553 (anti-CD 40) and 2C4 antibodies. In some embodiments, the antibodies are intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, or antibody fragments so long as they exhibit the desired biological activity.
For the treatment of Acute Myeloid Leukemia (AML), the compounds of the present disclosure can be used in combination with standard leukemia therapies, particularly in combination with therapies used to treat AML. In particular, the compounds of the present disclosure may be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for treating AML, such as daunorubicin, Adriamycin (Adriamycin), Ara-C, VP-16, teniposide, mitoxantrone, idamycin, carboplatin, and PKC 412.
Other anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog which is a 2' - α -hydroxyribose (arabinoside) derivative of deoxycytidine. Purine analogs of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate (fluorabane phosphate) are also included. Compounds that target, decrease or inhibit the activity of Histone Deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of enzymes known as histone deacetylases. Specific HDAC inhibitors include compounds disclosed in MS275, SAHA, FK228 (formerly FR901228), trichostatin a (trichostatin a) and US 6,552,065, including, but not limited to, N-hydroxy-3- [4- [ [ [2- (2-methyl-1H-indol-3-yl) -ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof, and N-hydroxy-3- [4- [ (2-hydroxyethyl) {2- (1H-indol-3-yl) ethyl ] -amino ] methyl ] phenyl ] -2E-2-acrylamide or a pharmaceutically acceptable salt thereof, especially lactate. In some embodiments, the somatostatin receptor antagonist is a compound that targets, treats, or inhibits a somatostatin receptor, such as octreotide (octreotide) and SOM 230. In some embodiments, the tumor cell destruction method is a method such as ionizing radiation. In some embodiments, the ionizing radiation is ionizing radiation that is carried out in the form of electromagnetic rays (e.g., X-rays and gamma rays) or particles (e.g., alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiotherapy and is known in the art. See Hellman, Principles of Cancer radiotherapy (Cancer), Oncology Principles and practices (Principles and Practice of Oncology), Devita et al, 4 th edition, Vol.1, p.248-275 (1993).
EDG binding agents and ribonucleotide reductase inhibitors are also included. In some embodiments, EDG binding agents are a class of immunosuppressive agents that modulate lymphocyte recirculation, such as FTY 720. In some embodiments, the ribonucleotide reductase inhibitor is a pyrimidine or purine nucleoside analog, including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C for ALL), and/or pentostatin (pentostatin). Ribonucleotide reductase inhibitors are in particular hydroxyurea or 2-hydroxy-1H-isoindole-1, 3-dione derivatives.
Those compounds, proteins or monoclonal antibodies which also comprise VEGF, inter alia, such as 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine or a pharmaceutically acceptable salt thereof, 1- (4-chloroanilino) -4- (4-pyridylmethyl) phthalazine succinate; angiostatinTM;EndostatinTM(ii) a Anthranilic acid amides; ZD 4190; ZD 6474; SU 5416; SU 6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab, VEGF aptamers, such as the marijuana supply (Macugon); FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2IgGI antibodies, Amphibin (Angiozyme) (RPI 4610) and bevacizumab (Avastin) TM)。
In some embodiments, photodynamic therapy is a therapy for treating or preventing cancer using certain chemicals known as photoactive compounds. Examples of photodynamic therapy include treatment with, for example, VisudyneTMAnd porfimer sodium.
In some embodiments, the angiogenesis inhibiting steroid is a compound that blocks or inhibits angiogenesis, such as anecortave (anecortave), triamcinolone (triamcinolone), hydrocortisone, 11-alpha-epihydrocortisone, deoxycorticosterol (cortixolone), 17 alpha-hydroxyprogesterone, corticosterone (corticosterone), deoxycorticosterone (desoxycorticosterone), testosterone, estrone, and dexamethasone.
In some embodiments, the corticosteroid-containing implant is a compound such as fluocinolone (fluocinolone) and dexamethasone.
Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, such as lymphokines or interferons; an antisense oligonucleotide or oligonucleotide derivative; shRNA or siRNA; or hybrid compounds or compounds with other or unknown mechanisms of action.
In some embodiments, the compounds of the present disclosure are also useful as adjunctive therapeutic compounds for use in combination with other drug substances, e.g., anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly for the treatment of obstructive or inflammatory tracheal diseases such as those mentioned above, e.g., as potentiators of therapeutic activity of the drug or as methods of reducing required dosages or potential side effects of the drug. In some embodiments, the disclosed compounds may be mixed with other drug substances in the form of a fixed pharmaceutical composition or they may be administered separately, before, simultaneously or after the other drug substances. Accordingly, the present disclosure provides a combination of a provided compound as described above and an anti-inflammatory, bronchodilatory, antihistamine or antitussive drug substance, the pharmaceutical composition of the provided compound and the drug substance being the same or different.
Suitable anti-inflammatory agents include steroids, especially glucocorticosteroids such as budesonide, beclomethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furoate; a non-steroidal glucocorticoid receptor agonist; LTB4 antagonists, such as LY293111, CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247; LTD4 antagonists, such as montelukast (montelukast) and zafirlukast (zafirlukast); PDE4 inhibitors, such as cilomilast (cilomilast) (II)
Figure BDA0003526174380002161
Glassware Sc), Roflumilast (Roflumilast) (Byk Gulden), V-11294A (Napp), BAY19-8004 (Bayer), SCH-351591 (Schering-Plough), Arofylline (Arofylline) (Almirall Prodesfara), PD189659/PD168787 (Pake-Davis), AWD-12-281 (Asta Medica)), CDC-801 (Seill gene), SeICID (TM) CC-10004 (Seal gene), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (synergic and cozeb), Rofluvilast (Byk Gulden), V-11294A (Napp), BAY19-8004 (Bayer Prodesr), PD 1899/PD 168787 (Pax-Dai), and K-4490 (Cox and Nippon-K)Industry (Kyowa Hakko Kogyo)); a2a agonist; an A2b antagonist; and β -2 adrenoceptor agonists such as salbutamol (salbutamol), metaproterenol, terbutaline, salmeterol, fenoterol, procaterol (procaterol) and especially formoterol and pharmaceutically acceptable salts thereof. Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, especially ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226(Chiesi) and glycopyrrolate.
Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophenol fumarate, clemastine fumarate, promethazine, loratadine (loratidine), desloratadine (desloratidine), diphenhydramine (diphenhydramine) and phenafenadine hydrochloride (fexofenadine hydrochloride), avastine (activivastine), astemizole (astemizole), azelastine (azelastine), ebastine (ebastine), epinastine (epinasstine), mizolastine (mizolastine) and terfenadine (tefenadine).
Other suitable combinations of compounds with anti-inflammatory agents are antagonists of chemokine receptors, for example CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, especially CCR-5 antagonists, such as the pioneer and bayan antagonists SC-351125, SCH-55700 and SCH-D, and Wutian antagonists, such as those combinations of N- [ [4- [ [ [6, 7-dihydro-2- (4-methylphenyl) -5H-benzo-cyclohept-8-yl ] carbonyl ] amino ] phenyl ] -methyl ] tetrahydro-N, N-dimethyl-2H-pyran-4-ammonium chloride (TAK-770).
The structures of active compounds identified by code number, common name or trade name can be obtained from The "Merck Index" of The current edition of The standard schema or from databases, such as The International patent (Patents) (e.g. The IMS World Publications).
The disclosed compounds may also be used in combination with known therapeutic methods, such as administration of hormones or radiation. In certain embodiments, provided compounds are used as radiosensitizers, particularly for treating tumors that exhibit poor sensitivity to radiotherapy.
The disclosed compounds may be administered alone or in combination with one or more other therapeutic compounds, with possible combination therapies employing fixed combinations or staggered or independent of each other to provide administration of the disclosed compounds and one or more other therapeutic compounds, or a combination of a fixed combination and one or more other therapeutic compounds. The disclosed compounds may be administered additionally or alternatively, especially in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention, or a combination of these, for the treatment of tumors. In the case of other treatment strategies, long-term therapy and adjuvant therapy are likewise possible, as described above. Other possible treatments are therapies that maintain the patient's state after tumor regression or even chemopreventive therapies (e.g. for patients at risk).
In some embodiments, the composition should be formulated such that a dose of between 0.01 and 100mg of compound per kg of body weight per day can be administered.
In some embodiments, in a composition comprising an additional therapeutic agent, the additional therapeutic agent and a compound of the present disclosure may act synergistically. In some embodiments, the amount of additional therapeutic agent will be less than that required in a monotherapy utilizing only the therapeutic agent. In such compositions, additional therapeutic agents may be administered at doses between 0.01 and 1,000^ g per kilogram of body weight per day.
The compounds of the present disclosure or pharmaceutical compositions thereof may also be incorporated into compositions for coating implantable medical devices, such as prostheses, prosthetic valves, vascular prostheses, stents, and catheters. Vascular stents, for example, have been used to overcome restenosis (restenosis of the vessel wall after injury). However, patients using stents or other implantable devices are at risk of forming clots or platelet activation. These undesirable effects can be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a compound of the present disclosure. Implantable devices coated with the disclosed compounds are another embodiment of the disclosure.
Further, the present disclosure provides the following embodiments:
1. a medicament, comprising:
(ii) an antibody-binding moiety,
A target binding moiety, and
the optional presence of a linker moiety or moieties,
wherein the target binding moiety specifically binds to CD 38.
2. The agent of embodiment 1, wherein the agent has the structure of formula I:
Figure BDA0003526174380002181
or a pharmaceutically acceptable salt thereof, wherein:
each of a and b is independently 1 to 200;
each ABT is independently an antibody binding moiety;
l is a linker moiety connecting ABT and TBT; and is
Each TBT is independently a target binding moiety.
3. The agent of embodiment 1, wherein the agent has the structure:
Figure BDA0003526174380002191
or a pharmaceutically acceptable salt thereof, wherein:
each of a and b is independently 1 to 200;
each ABT is independently an antibody binding moiety;
l is a bivalent linker moiety linking ABT to TBT;
each Xaa is independently a residue of an amino acid or amino acid analog;
y is 5 to 20;
LTa linker moiety which is two separate residues of an amino acid or amino acid analogue and is independently a covalent bond, or is selected from C1-C6Aliphatic radicals or having 1 to 5 hetero atomsC1-C6A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R') 2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each RcIndependently is-La-R′;
t is 0 to 50;
each LaIndependently a covalent bond, or is selected from C1-C50Aliphatic radical or C having 1 to 5 hetero atoms1-C50A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from C3-20Cycloaliphatic Ring, C6-20An aryl ring, a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms, and a 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30Aliphatic radical, C having 1 to 10 heteroatoms1-30Heteroaliphatic radical, C6-30Aryl radical, C6-30Arylaliphatic radical, C having 1 to 10 heteroatoms6-30Aryl heteroaliphatic, 5-to 30-membered heteroaryl having 1 to 10 heteroatoms and 3-to 30-membered heterocyclyl having 1 to 10 heteroatoms, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, together with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the atom; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the intervening atoms.
4. The medicament according to any one of the preceding embodiments, wherein a is 1.
5. The medicament according to any one of the preceding embodiments, wherein b is 1.
6. The agent according to any one of the preceding embodiments, wherein the target binding moiety is or comprises (Xaa) y, wherein each Xaa is independently a residue of an amino acid or amino acid analog, and y is 5 to 20.
7. A medicament, comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
the optional presence of a linker moiety or moieties,
wherein the target binding moiety has the following structure:
Figure BDA0003526174380002201
or a salt thereof, wherein:
each Xaa is independently a residue of an amino acid or amino acid analog;
y is 5 to 20;
LTa linker moiety which is two separate residues of an amino acid or amino acid analogue and is independently a covalent bond, or is selected from C1-C6Aliphatic radical or C having 1 to 5 hetero atoms1-C6A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R') 2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each RcIndependently is-La-R′;
t is 0 to 50;
each LaIndependently a covalent bond, or is selected from C1-C50Aliphatic radical or C having 1 to 5 hetero atoms1-C50A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from C3-20Cycloaliphatic Ring, C6-20An aryl ring, a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms, and a 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30Aliphatic radical, C having 1 to 10 heteroatoms1-30Heteroaliphatic radical, C6-30Aryl radical, C6-30Arylaliphatic radical, C having 1 to 10 heteroatoms6-30Aryl heteroaliphatic, 5-to 30-membered heteroaryl having 1 to 10 heteroatoms and 3-to 30-membered heterocyclyl having 1 to 10 heteroatoms, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, together with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the atom; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the intervening atoms.
8. The agent of any one of the preceding embodiments, wherein the antibody binding moiety can bind to two or more antibodies having different Fab regions.
9. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety can bind to an Fc region.
10. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises optionally substituted
Figure BDA0003526174380002211
11. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002212
12. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises optionally substituted
Figure BDA0003526174380002213
13. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002221
14. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises optionally substituted
Figure BDA0003526174380002222
15. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002223
16. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises optionally substituted
Figure BDA0003526174380002224
17. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002225
18. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises one or more amino acid residues.
19. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises a peptide moiety.
20. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises Rc- (Xaa) z-or
Figure BDA0003526174380002226
Or a salt form thereof.
21. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002231
22. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002232
23. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002233
24. The agent according to any one of the preceding embodiments, wherein the antibody binding moiety is or comprises
Figure BDA0003526174380002241
25. The agent of any one of embodiments 1-9, wherein the antibody binding moiety has R c- (Xaa) z-or
Figure BDA0003526174380002242
Or a salt form thereof.
26. The agent of any one of embodiments 1-9 and 25, wherein the antibody binding portion has a structure of DCAWHLGELVWCT or a salt form thereof, wherein the two C residues are linked by-S-.
27. The medicament according to any one of the preceding embodiments, wherein- (Xaa) y-comprises:
-XaaT1-XaaT2-(Xaa)y′-XaaT3-XaaT4-XaaT5-,
wherein:
y' is 0 to 8;
XaaT1for C whose side chain is substituted1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT2including an optionally substituted aromatic group for its side chain or being optionally substituted C3-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT3is C whose side chain is optionally substituted2-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT4including an optionally substituted aromatic group for its side chain or being optionally substituted C3-C8A residue of an amino acid or amino acid analog of an aliphatic group; and is
XaaT5For C whose side chain is substituted1-C8Aliphatic amino acids or amino acidsA residue of an analog.
28. The agent of embodiment 27, wherein y' is 4.
29. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is C whose side chain is optionally substituted2-C8A residue of an alkyl group.
30. The medicament according to any one of embodiments 27 to 28, wherein XaaT1For its side chain being unsubstituted, linear C2-C8A residue of an alkyl group.
31. The medicament according to any one of embodiments 27 to 28, wherein XaaT1For its side chain being unsubstituted, linear C2-C8A residue of an alkyl group.
32. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is a residue whose side chain is n-pentyl.
33. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of Ahp, Y, W, S, K or K (MePEG4 c).
34. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of Ahp.
35. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of Y.
36. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of W.
37. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of S.
38. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of K.
39. The medicament according to any one of embodiments 27 to 28, wherein XaaT1Is the residue of K (MePEG4 c).
40. The medicament according to any one of embodiments 27 to 38, wherein Xaa T2For its side chain is-CH2-the residue of R, wherein R is optionally substituted phenyl.
41. According to any one of embodiments 27 to 38The medicament of, wherein XaaT2Is C whose side chain is optionally substituted3-C8A residue of an aliphatic group.
42. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is the residue of Y, W, Ahp, Bph, L or A.
43. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is the residue of Y.
44. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is the residue of W.
45. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is the residue of Ahp.
46. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is the residue of Bph.
47. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is a residue of L.
48. The medicament according to any one of embodiments 27 to 38, wherein XaaT2Is the residue of A.
49. The medicament according to any one of embodiments 27 to 48, wherein XaaT3Is a residue of L, Ahp, V, T, Hse or MetO 2.
50. The medicament according to any one of embodiments 27 to 48, wherein XaaT3Is a residue of L.
51. The medicament according to any one of embodiments 27 to 48, wherein Xaa T3Is the residue of Ahp.
52. The medicament according to any one of embodiments 27 to 48, wherein XaaT3Is the residue of V.
53. The medicament according to any one of embodiments 27 to 48, wherein XaaT3Is the residue of T.
54. The medicament according to any one of embodiments 27 to 48, wherein XaaT3Is the residue of Hse.
55. The medicament according to any one of embodiments 27 to 48, wherein XaaT3Is the residue of MetO 2.
56. According to implementationThe medicament of any one of embodiments 27 to 55, wherein XaaT4Is a residue whose side chain includes an optionally substituted aromatic group.
57. The medicament according to any one of embodiments 27 to 55, wherein XaaT4For its side chain is-CH2-the residue of R, wherein R is optionally substituted phenyl.
58. The medicament according to any one of embodiments 27 to 55, wherein XaaT4Including optionally substituted C for its side chain3-C8A residue of an aliphatic group.
59. The medicament according to any one of embodiments 27 to 55, wherein XaaT4Residues of Bph, V or Ahp.
60. The medicament according to any one of embodiments 27 to 55, wherein XaaT4Is the residue of Bph.
61. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is C whose side chain is optionally substituted 2-C6A residue of an alkyl group.
62. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is C whose side chain is optionally substituted2-C6A linear alkyl residue.
63. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is a residue whose side chain is n-pentyl.
64. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Residues of Ahp, Bph, Ado, Ano, PhNle or PhNva.
65. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is the residue of Ahp.
66. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is the residue of Bph.
67. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is the residue of Ado.
68. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is a residue of Ano.
69. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is a residue of PhNle.
70. The medicament according to any one of embodiments 27 to 60, wherein XaaT5Is a residue of PhNva.
71. The agent of any one of embodiments 27 to 70, wherein- (Xaa) y-is or comprises:
-(Xaa)a1-(Xaa)a2-(Xaa)a3-(Xaa)a4-(Xaa)a5-(Xaa)a6-(Xaa)a7-(Xaa)a8-(Xaa)a9-(Xaa)a10-(Xaa)a11-(Xaa)a12-(Xaa)a13-,
wherein:
each of a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and a13 is independently 0 to 5;
(Xaa)a3Is or comprises XaaT1
(Xaa)a4Is or comprises XaaT2
(Xaa)a9Is or comprises XaaT3
(Xaa)a10Is or comprises XaaT4(ii) a And is
(Xaa)a11Is or comprises XaaT5
72. The medicament according to any one of embodiments 27 to 71, wherein (Xaa)a1Is or includes A, K or K (MePEG4 c).
73. The medicament according to any one of embodiments 27 to 71, wherein (Xaa)a1Wherein a1 is 1 and Xaa is the residue of A.
74. The medicament according to any one of embodiments 27 to 71, wherein (Xaa)a1Wherein a1 is 1 and Xaa is a residue of K.
75. The medicament according to any one of embodiments 27 to 71, wherein (Xaa)a1In (a 1) is 1 and Xaa is the residue of K (MePEG4 c).
76. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or comprises R, S, D, Y, A, W, K, 4Py2NH2, Cit, F3G,hCit, K (MePEG4c), RNdMe, RNMe or RNNdMe.
77. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2In (a 2) is 1 and Xaa is a residue of R, S, D, Y, W, A or S.
78. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2In (a 2) is 1 and Xaa is a residue of R, S, D, Y, W, A or S.
79. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes R.
80. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes S.
81. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes D.
82. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes Y.
83. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes A.
84. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes W.
85. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes K.
86. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes 4Py2NH 2.
87. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes Cit.
88. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes F3G.
89. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes hCit.
90. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes K (MePEG4 c).
91. According to examples 27 to75 wherein (Xaa)a2Is or includes RNdMe.
92. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Is or includes RNMe.
93. The medicament according to any one of embodiments 27 to 75, wherein (Xaa)a2Wherein a2 is 1 and Xaa is the residue of RNNdMe.
94. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Is or includes H, Y, S, L, A or W6N.
95. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Wherein a5 is 1 and Xaa is a residue of H, Y, S, L, A or W.
96. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of H or Y.
97. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of H.
98. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of Y.
99. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of S.
100. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of L.
101. The medicament according to any one of embodiments 27 to 93, wherein (Xaa) a5Wherein a5 is 1 and Xaa is the residue of A.
102. The medicament according to any one of embodiments 27 to 93, wherein (Xaa)a5In (a 5) is 1 and Xaa is the residue of W6N.
103. The medicament according to any one of embodiments 27 to 102, wherein (Xaa)a6Is or includes D, G, R, Y, H, W, A or Y.
104. The medicament according to any one of embodiments 27 to 102, wherein (Xaa)a6Wherein a6 is 1 and Xaa is D, G, R, Y, H, W, A or Y.
105. The medicament according to any one of embodiments 27 to 102, wherein (Xaa)a6Wherein a6 is 1 and Xaa is D, G or R.
106. The medicament according to any one of embodiments 27 to 102, wherein (Xaa)a6Wherein a6 is 1 and Xaa is the residue of D.
107. The medicament according to any one of embodiments 27 to 102, wherein (Xaa)a6Wherein a6 is 1 and Xaa is the residue of A.
108. The agent of any one of embodiments 27 to 107, wherein (Xaa)a7Is or includes G, D, E, Q, N, R, MetO2, S, Har or A.
109. The agent of any one of embodiments 27 to 107, wherein (Xaa)a7Is or includes G, D, E, Q or N.
110. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7Wherein a7 is 1 and Xaa is the residue of G.
111. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7Wherein a7 is 1 and Xaa is the residue of D, G or A.
112. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7Wherein a7 is 1 and Xaa is the residue of D.
113. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7Wherein a7 is 1 and Xaa is the residue of E.
114. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7Wherein a7 is 1 and Xaa is the residue of N.
115. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7Wherein a7 is 1 and Xaa is the residue of Q.
116. The medicament according to any one of embodiments 27 to 107, wherein (Xaa)a7In (a 7) is 1 and Xaa is the residue of MetO 2.
117. The agent of any one of embodiments 27-107, wherein at (X)aa)a7Wherein a7 is 1 and Xaa is the residue of A.
118. The medicament according to any one of embodiments 27 to 117, wherein (Xaa)a8Is or includes V, A, D, G, W, S or T.
119. The medicament according to any one of embodiments 27 to 117, wherein (Xaa)a8In (a 8) is 1 and Xaa is the residue of V, A, D, G, W, S or T.
120. The medicament according to any one of embodiments 27 to 117, wherein (Xaa) a8Wherein a8 is 1 and Xaa is the residue of V.
121. The medicament according to any one of embodiments 27 to 117, wherein (Xaa)a8Wherein a8 is 1 and Xaa is the residue of A.
122. The medicament according to any one of embodiments 27 to 121, wherein (Xaa)a12Is or includes D, A, S, G or Ahp.
123. The medicament according to any one of embodiments 27 to 121, wherein (Xaa)a12Wherein a12 is 1 and Xaa is D, S, G or Ahp.
124. The medicament according to any one of embodiments 27 to 121, wherein (Xaa)a12Wherein a12 is 1 and Xaa is the residue of D.
125. The medicament according to any one of embodiments 27 to 121, wherein (Xaa)a12Wherein a12 is 1 and Xaa is the residue of A.
126. The medicament according to any one of embodiments 27 to 125, wherein (Xaa)a13Is or includes C.
127. The medicament according to any one of embodiments 27 to 125, wherein (Xaa)a13Wherein a13 is 1 and Xaa is a residue of C.
128. The agent of any one of embodiments 1 to 26, wherein- (Xaa) y-comprises:
-XaaT6-(Xaa)y′-XaaT7-XaaT8-XaaT9-XaaT10-XaaT11-,
wherein:
y' is 0 to 8;
XaaT6is its side chain isSubstituted C1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT7is C whose side chain is optionally substituted2-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT8Is a residue of proline or an amino acid analogue thereof;
XaaT9including an optionally substituted aromatic group for its side chain or being optionally substituted C1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT10for C whose side chain is substituted1-C8A residue of an aliphatic group amino acid or an amino acid analog or a residue of an amino acid in which an amino group thereof is substituted; and is
XaaT11Including an optionally substituted aromatic group for its side chain or being optionally substituted C1-C8Aliphatic amino acids or amino acid analogues.
129. The agent of embodiment 128, wherein y' is 1.
130. The medicament according to any one of embodiments 128 to 129, wherein XaaT6Side chain of (A) is-CH2-R, wherein R is optionally substituted phenyl.
131. The medicament according to any one of embodiments 128 to 130, wherein XaaT6Is an amino acid residue whose amino group has the structure of-N (R) -wherein R is optionally substituted C1-6An aliphatic group.
132. The agent of any one of embodiments 128 to 131 wherein XaaT6An amino acid residue whose amino group has the structure of-N (Me) -.
133. The medicament according to any one of embodiments 128 to 129, wherein XaaT6Is a residue of MeF, L or S.
134. The medicament according to any one of embodiments 128 to 129, wherein Xaa T6Is the residue of MeF.
135. The medicament according to any one of embodiments 128 to 134, wherein XaaT7Is a residue of L or MeF.
136. The medicament according to any one of embodiments 128 to 134, wherein XaaT7Is a residue of L.
137. The medicament according to any one of embodiments 128 to 136, wherein XaaT8Is the residue of P.
138. The medicament according to any one of embodiments 128 to 137, wherein XaaT9Is a residue whose side chain includes an optionally substituted aromatic group.
139. The medicament according to any one of embodiments 128 to 138, wherein XaaT9For its side chain is-CH2-the residue of R, wherein R is optionally substituted phenyl.
140. The medicament according to any one of embodiments 128 to 137, wherein XaaT9Is C whose side chain is optionally substituted1-C8A residue of an aliphatic group.
141. The medicament according to any one of embodiments 128 to 137, wherein XaaT9Is a residue of Bph, D or S.
142. The medicament according to any one of embodiments 128 to 137, wherein XaaT9Is the residue of Bph.
143. The medicament according to any one of embodiments 128 to 142, wherein XaaT10For C whose side chain is substituted1-C8A residue of an aliphatic group.
144. The medicament according to any one of embodiments 128 to 142, wherein Xaa T10Is a residue of V or L.
145. The medicament according to any one of embodiments 128 to 142, wherein XaaT10Is the residue of V.
146. The medicament according to any one of embodiments 128 to 142, wherein XaaT10Is a residue including a substituted amino group.
147. The medicament according to any one of embodiments 128 to 142, wherein XaaT10Is the residue of MeG.
148. The medicament according to any one of embodiments 128 to 147, wherein XaaT11For the side chain thereof including optionally substitutedA residue of a substituted aromatic group.
149. The medicament according to any one of embodiments 128 to 147, wherein XaaT11For its side chain is-CH2-the residue of R, wherein R is optionally substituted aryl or heteroaryl.
150. The medicament according to any one of embodiments 128 to 147, wherein XaaT11Is the residue of W.
151. The medicament according to any one of embodiments 128 to 147, wherein XaaT11Is C whose side chain is optionally substituted1-C8A residue of an aliphatic group.
152. The medicament according to any one of embodiments 128 to 147, wherein XaaT11Is the residue of R.
153. The agent of any one of embodiments 128 to 152, wherein- (Xaa) y-is or comprises:
-(Xaa)a1-(Xaa)a2-(Xaa)a3-(Xaa)a4-(Xaa)a5-(Xaa)a6-(Xaa)a7-(Xaa)a8-(Xaa)a9-(Xaa)a10-(Xaa)a11-(Xaa)a12-,
wherein:
each of a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and a12 is independently 0 to 5;
(Xaa)a4Is or comprises XaaT6
(Xaa)a6Is or comprises XaaT7
(Xaa)a7Is or comprises XaaT8
(Xaa)a8Is or comprises XaaT9
(Xaa)a9Is or comprises XaaT10(ii) a And is
(Xaa)a10Is or comprises XaaT11
154. The agent of any one of embodiments 128 to 153, wherein (Xaa)a1Is or includes A.
155. The agent of any one of embodiments 128 to 153, wherein (Xaa)a1Wherein a1 is 1 and Xaa is the residue of A.
156. The agent of any one of embodiments 128 to 155, wherein (Xaa)a2Is or includes L, A or P.
157. The agent of any one of embodiments 128 to 155 wherein (Xaa)a2Wherein a2 is 1 and Xaa is the residue of L.
158. The agent of any one of embodiments 128 to 155 wherein (Xaa)a2Wherein a2 is 1 and Xaa is the residue of A.
159. The agent of any one of embodiments 128 to 158, wherein (Xaa)a3Is or includes H, R or A.
160. The agent of any one of embodiments 128 to 158, wherein (Xaa)a3Wherein a3 is 1 and Xaa is the residue of H, R or A.
161. The agent of any one of embodiments 128 to 158, wherein (Xaa)a3Wherein a3 is 1 and Xaa is the residue of H, R or A.
162. The agent of any one of embodiments 128 to 158, wherein (Xaa)a3Wherein a3 is 1 and Xaa is the residue of H.
163. The agent of any one of embodiments 128 to 158, wherein (Xaa)a3Wherein a3 is 1 and Xaa is the residue of A.
164. The agent of any one of embodiments 128 to 163, wherein (Xaa)a5Is or includes V, A or MeG.
165. The medicament according to any one of embodiments 128 to 163, wherein (Xaa)a5In (a 5) is 1 and Xaa is the residue of V, A or MeG.
166. The medicament according to any one of embodiments 128 to 163, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of V.
167. The medicament according to any one of embodiments 128 to 163, wherein (Xaa)a5Wherein a5 is 1 and Xaa is the residue of A.
168. The agent of any one of embodiments 128 to 167, wherein (Xaa)a11Is or includes V, A, D or MeG.
169. The agent of any one of embodiments 128 to 167, wherein (Xaa)a11In (a 11) is 1 and Xaa is the residue of V, A, D or MeG.
170. The agent of any one of embodiments 128 to 167, wherein (Xaa)a11Wherein a11 is 1 and Xaa is the residue of V.
171. The agent of any one of embodiments 128 to 167, wherein (Xaa)a11Wherein a11 is 1 and Xaa is the residue of A.
172. The agent of any one of embodiments 128 to 167, wherein (Xaa) a12Is or includes C.
173. The agent of any one of embodiments 128 to 167, wherein (Xaa)a12Wherein a12 is 1 and Xaa is a residue of C.
174. The medicament of any one of embodiments 27 to 173, wherein two Xaa are linked together.
175. The agent of any one of embodiments 27 to 174, wherein two Xaa are via a c- (o) -CH2-the linkers of the structure are linked together.
176. The agent of embodiment 175, wherein-c (o) -amino bonded to Xaa.
177. The agent of embodiment 176, wherein Xaa is the N-terminal residue.
178. The agent of any one of embodiments 175-177, wherein-CH2-S-bonded to the side chain of Xaa.
179. The agent of embodiment 178, wherein Xaa is a C-terminal residue.
180. The agent of embodiment 178 or 179, wherein Xaa is C.
181. The agent of any one of embodiments 1-127 and 174-180, wherein the target binding moiety or
Figure BDA0003526174380002331
Is or comprise
Figure BDA0003526174380002332
Or a salt form thereof.
182. The agent of any one of embodiments 1-127 and 174-180, wherein the target binding moiety or
Figure BDA0003526174380002341
Is or comprise
Figure BDA0003526174380002342
Or a salt form thereof.
183. The agent of any one of embodiments 1-127 and 174-180, wherein the target binding moiety or
Figure BDA0003526174380002343
Is or comprise
Figure BDA0003526174380002344
Or a salt form thereof.
184. The agent of any one of embodiments 1-127 and 174-180, wherein the target binding moiety or
Figure BDA0003526174380002351
Is or comprise
Figure BDA0003526174380002352
Or a salt form thereof.
185. The agent of any one of embodiments 1-127 and 174-180, wherein the target binding moiety or
Figure BDA0003526174380002353
Is or comprise
Figure BDA0003526174380002354
Or a salt form thereof.
186. The agent of any one of embodiments 1-26 and 128-180, wherein the target binding moiety or
Figure BDA0003526174380002361
Is or comprise
Figure BDA0003526174380002362
Or a salt form thereof.
187. The agent of any one of embodiments 1-26 and 128-180, wherein the target binding moiety or
Figure BDA0003526174380002363
Is or comprise
Figure BDA0003526174380002364
Or a salt form thereof.
188. The agent according to any one of embodiments 1 to 26, wherein the target binding moiety or- (Xaa) y-is or comprises a peptide of:
(1) a polypeptide having an amino acid sequence represented by any one of SEQ ID nos. 1 to 34:
(2) a polypeptide having an amino acid sequence represented by any one of SEQ ID nos. 1 to 34, wherein the amino acid residue at the N-terminus is chloroacetylated (e.g., at the amino group thereof);
(3) a polypeptide having an amino acid sequence with a deletion, addition, substitution or insertion of one or more amino acids in any one of SEQ ID nos. 1-34, which does not include a deleted amino acid sequence having a Cys at the C-terminus in SEQ ID nos. 1-34;
(4) A polypeptide having an amino acid sequence represented by any one of SEQ ID nos. 1-34 having a deletion, addition, substitution, or insertion of one or more amino acids in any one of SEQ ID nos. 1-34, excluding a deleted amino acid sequence having a Cys at the C-terminus in any one of SEQ ID nos. 1-34, wherein the amino acid at the N-terminus is chloroacetylated (e.g., at its amino acid); or
(5) The polypeptide according to any one of (1) to (4) above, wherein the polypeptide has a cyclized structure.
189. The agent according to any one of embodiments 1 to 26, wherein the target binding moiety or- (Xaa) y-is or comprises a peptide of:
(1) a polypeptide having an amino acid sequence represented by SEQ ID No.1 or 2:
Ala Arg Ahp Tyr His Asp Gly Val Leu Bph Ahp Asp Cys(SEQ ID NO.1),
Ala Leu His MePhe Val Leu Pro Bph Val Trp Val Cys(SEQ ID NO.2);
(2) a polypeptide having an amino acid sequence represented by SEQ ID No.1 or 2, wherein Ala at the N-terminus is chloroacetylated Ala;
(3) a polypeptide having a deletion, addition, substitution, or insertion of one or more amino acids in SEQ ID No.1 or 2 in the amino acid sequence, which does not include an amino acid sequence having a deletion of Cys at the C-terminus in SEQ ID No.1 or 2;
(4) a polypeptide having an amino acid sequence represented by SEQ ID No.1 or 2, wherein Ala at the N-terminus is chloroacetylated Ala, having a deletion, addition, substitution, or insertion of one or more amino acids in SEQ ID No.1 or 2, which excludes a deleted amino acid sequence having Cys at the C-terminus in SEQ ID No.1 or 2; or
(5) The polypeptide according to any one of (1) to (4) above, wherein the polypeptide has a cyclized structure.
190. The agent according to any one of embodiments 1 to 26 wherein the binding moiety of interest or- (Xaa) y-is or includes a peptide having an amino acid sequence of any one of SEQ ID No. 1-34.
191. The agent of any one of embodiments 188 to 190, wherein the target binding moiety or- (Xaa) y-has a cyclized structure.
192. The agent of any one of embodiments 1 to 26, wherein the target binding moiety is derived from, or
Figure BDA0003526174380002371
The following are: a structure selected from S-1 to S-39 or a pharmaceutically acceptable salt thereof.
193. The medicament of embodiment 192, wherein the structure is S-1 or a pharmaceutically acceptable salt thereof.
194. The medicament of embodiment 192, wherein the structure is S-2 or a pharmaceutically acceptable salt thereof.
195. The medicament of embodiment 192, wherein the structure is S-3 or a pharmaceutically acceptable salt thereof.
196. The medicament of embodiment 192, wherein the structure is S-4 or a pharmaceutically acceptable salt thereof.
197. The medicament of embodiment 192, wherein the structure is S-5 or a pharmaceutically acceptable salt thereof.
198. The medicament of embodiment 192, wherein the structure is S-6 or a pharmaceutically acceptable salt thereof.
199. The medicament of embodiment 192, wherein the structure is S-7 or a pharmaceutically acceptable salt thereof.
200. The medicament of embodiment 192, wherein the structure is S-8 or a pharmaceutically acceptable salt thereof.
201. The medicament of embodiment 192, wherein the structure is S-9 or a pharmaceutically acceptable salt thereof.
202. The medicament of embodiment 192, wherein the structure is S-10 or a pharmaceutically acceptable salt thereof.
203. The medicament of embodiment 192, wherein the structure is S-11 or a pharmaceutically acceptable salt thereof.
204. The medicament of embodiment 192, wherein the structure is S-12 or a pharmaceutically acceptable salt thereof.
205. The medicament of embodiment 192, wherein the structure is S-13 or a pharmaceutically acceptable salt thereof.
206. The medicament of embodiment 192, wherein the structure is S-14 or a pharmaceutically acceptable salt thereof.
207. The medicament of embodiment 192, wherein the structure is S-15 or a pharmaceutically acceptable salt thereof.
208. The medicament of embodiment 192, wherein the structure is S-16 or a pharmaceutically acceptable salt thereof.
209. The medicament of embodiment 192, wherein the structure is S-17 or a pharmaceutically acceptable salt thereof.
210. The medicament of embodiment 192, wherein the structure is S-18 or a pharmaceutically acceptable salt thereof.
211. The medicament of embodiment 192, wherein the structure is S-19 or a pharmaceutically acceptable salt thereof.
212. The medicament of embodiment 192, wherein the structure is S-20 or a pharmaceutically acceptable salt thereof.
213. The medicament of embodiment 192, wherein the structure is S-21 or a pharmaceutically acceptable salt thereof.
214. The medicament of embodiment 192, wherein the structure is S-22 or a pharmaceutically acceptable salt thereof.
215. The medicament of embodiment 192, wherein the structure is S-23 or a pharmaceutically acceptable salt thereof.
216. The medicament of embodiment 192, wherein the structure is S-24 or a pharmaceutically acceptable salt thereof.
217. The medicament of embodiment 192, wherein the structure is S-25 or a pharmaceutically acceptable salt thereof.
218. The medicament of embodiment 192, wherein the structure is S-26 or a pharmaceutically acceptable salt thereof.
219. The medicament of embodiment 192, wherein the structure is S-27 or a pharmaceutically acceptable salt thereof.
220. The medicament of embodiment 192, wherein the structure is S-28 or a pharmaceutically acceptable salt thereof.
221. The medicament of embodiment 192, wherein the structure is S-29 or a pharmaceutically acceptable salt thereof.
222. The medicament of embodiment 192, wherein the structure is S-30 or a pharmaceutically acceptable salt thereof.
223. The medicament of embodiment 192, wherein the structure is S-31 or a pharmaceutically acceptable salt thereof.
224. The medicament of embodiment 192, wherein the structure is S-32 or a pharmaceutically acceptable salt thereof.
225. The medicament of embodiment 192, wherein the structure is S-33 or a pharmaceutically acceptable salt thereof.
226. The medicament of embodiment 192, wherein the structure is S-34 or a pharmaceutically acceptable salt thereof.
227. The medicament of embodiment 192, wherein the structure is S-35 or a pharmaceutically acceptable salt thereof.
228. The medicament of embodiment 192, wherein the structure is S-36 or a pharmaceutically acceptable salt thereof.
229. The medicament of embodiment 192, wherein the structure is S-37 or a pharmaceutically acceptable salt thereof.
230. The medicament of embodiment 192, wherein the structure is S-38 or a pharmaceutically acceptable salt thereof.
231. The medicament of embodiment 192, wherein the structure is S-39 or a pharmaceutically acceptable salt thereof.
232. The medicament of any one of the preceding embodiments, comprising a linker.
233. The agent of any one of the preceding embodiments, wherein the linker is or comprises- (CH)2CH2O) n-where n is 1 to 20.
234. The agent of any one of the preceding embodiments, wherein the linker is or comprises- (CH)2CH2O) n-where n is about 5 to 20.
235. The agent of any one of the preceding embodiments, wherein the linker is or comprises- (CH)2CH2O) n-where n is about 10 to 20.
236. The agent of any one of the preceding embodiments, wherein the linker comprises one or more amino acid residues.
237. The agent of any one of the preceding embodiments, wherein the linker comprises one or more natural amino acid residues.
238. The agent of any one of the preceding embodiments, wherein the linker comprises one or more unnatural amino acid residues.
239. The agent of any one of the preceding embodiments, wherein the linker comprises one or more D-amino acid residues.
240. The agent of any one of the preceding embodiments, wherein the linker is or comprises a glycine residue.
241. The agent of any one of the preceding embodiments, wherein the linker is or comprises a β -alanine residue.
242. The agent of any one of the preceding embodiments, wherein the linker is or comprises a linker having-c (o) - (CH)2CH2O)n-CH2CH2NR' -structure or a salt form thereof, wherein n is 0 to 20.
243. The agent of any one of the preceding embodiments, wherein the linker is or comprises a linker having-c (o) - (CH)2CH2O)n-CH2CH2NR' -structure or a salt form thereof, wherein n is 0 to 12.
244. The agent of any one of embodiments 242-243, wherein R' is-H.
245. The agent of any one of the preceding embodiments, wherein the linker is or comprises-Gly-.
246. The agent of any one of the preceding embodiments, wherein the linker is or comprises
247. The agent of any one of the preceding embodiments, wherein the linker comprises a cycloaddition product moiety.
248. The medicament of any of the preceding embodiments, wherein the linker comprises
Figure BDA0003526174380002391
249. A medicament, wherein the medicament is I-3 or a pharmaceutically acceptable salt thereof.
250. A medicament, wherein the medicament is I-5 or a pharmaceutically acceptable salt thereof.
251. A pharmaceutical agent, wherein the pharmaceutical agent is I-6 or a pharmaceutically acceptable salt thereof.
252. A medicament, wherein the medicament is I-7 or a pharmaceutically acceptable salt thereof.
253. A medicament, wherein the medicament is I-8 or a pharmaceutically acceptable salt thereof.
254. A medicament, wherein the medicament is I-9 or a pharmaceutically acceptable salt thereof.
255. A pharmaceutical agent, wherein the pharmaceutical agent is I-10 or a pharmaceutically acceptable salt thereof.
256. A pharmaceutical agent, wherein the pharmaceutical agent is I-11 or a pharmaceutically acceptable salt thereof.
257. A pharmaceutical agent, wherein the pharmaceutical agent is I-12 or a pharmaceutically acceptable salt thereof.
258. A pharmaceutical agent, wherein the pharmaceutical agent is I-13 or a pharmaceutically acceptable salt thereof.
259. A medicament, wherein the medicament is I-15 or a pharmaceutically acceptable salt thereof.
260. A pharmaceutical agent, wherein the pharmaceutical agent is I-16 or a pharmaceutically acceptable salt thereof.
261. A medicament, wherein the medicament is I-17 or a pharmaceutically acceptable salt thereof.
262. A pharmaceutical agent, wherein the pharmaceutical agent is I-19 or a pharmaceutically acceptable salt thereof.
263. A pharmaceutical agent, wherein the pharmaceutical agent is I-24 or a pharmaceutically acceptable salt thereof.
264. The medicament of any one of embodiments 248-263, wherein
Figure BDA0003526174380002401
Is composed of
Figure BDA0003526174380002402
265. The medicament of any one of embodiments 248-264, wherein
Figure BDA0003526174380002403
Is composed of
Figure BDA0003526174380002404
266. A medicament, wherein the medicament is I-1 or a pharmaceutically acceptable salt thereof.
267. A medicament, wherein the medicament is I-2 or a pharmaceutically acceptable salt thereof.
268. A pharmaceutical agent, wherein the pharmaceutical agent is I-4 or a pharmaceutically acceptable salt thereof.
269. A pharmaceutical agent, wherein the pharmaceutical agent is I-14 or a pharmaceutically acceptable salt thereof.
270. A pharmaceutical agent, wherein the pharmaceutical agent is I-18 or a pharmaceutically acceptable salt thereof.
271. A medicament, wherein the medicament is I-25 or a pharmaceutically acceptable salt thereof.
272. A pharmaceutical agent, wherein the pharmaceutical agent is I-26 or a pharmaceutically acceptable salt thereof.
273. A pharmaceutical agent, wherein the pharmaceutical agent is I-27 or a pharmaceutically acceptable salt thereof.
274. A pharmaceutical agent, wherein the pharmaceutical agent is I-28 or a pharmaceutically acceptable salt thereof.
275. A pharmaceutical agent, wherein the pharmaceutical agent is I-29 or a pharmaceutically acceptable salt thereof.
276. A pharmaceutical agent, wherein the pharmaceutical agent is I-30 or a pharmaceutically acceptable salt thereof.
277. A pharmaceutical agent, wherein the pharmaceutical agent is I-31 or a pharmaceutically acceptable salt thereof.
278. A pharmaceutical agent, wherein the pharmaceutical agent is I-32 or a pharmaceutically acceptable salt thereof.
279. An agent, wherein the agent is I-33 or a pharmaceutically acceptable salt thereof.
280. A pharmaceutical agent, wherein the pharmaceutical agent is I-34 or a pharmaceutically acceptable salt thereof.
281. A pharmaceutical agent, wherein the pharmaceutical agent is I-35 or a pharmaceutically acceptable salt thereof.
282. A pharmaceutical agent, wherein the pharmaceutical agent is I-36 or a pharmaceutically acceptable salt thereof.
283. An agent, wherein the agent is I-37 or a pharmaceutically acceptable salt thereof.
284. A pharmaceutical agent, wherein the pharmaceutical agent is I-38 or a pharmaceutically acceptable salt thereof.
285. A medicament, wherein the medicament is I-39 or a pharmaceutically acceptable salt thereof.
286. A pharmaceutical agent, wherein the pharmaceutical agent is I-40 or a pharmaceutically acceptable salt thereof.
287. A pharmaceutical agent, wherein the pharmaceutical agent is I-41 or a pharmaceutically acceptable salt thereof.
288. A pharmaceutical agent, wherein the pharmaceutical agent is I-42 or a pharmaceutically acceptable salt thereof.
289. A pharmaceutical agent, wherein the pharmaceutical agent is I-43 or a pharmaceutically acceptable salt thereof.
290. A pharmaceutical agent, wherein the pharmaceutical agent is I-44 or a pharmaceutically acceptable salt thereof.
291. A medicament, wherein the medicament is I-45 or a pharmaceutically acceptable salt thereof.
292. A pharmaceutical agent, wherein the pharmaceutical agent is I-46 or a pharmaceutically acceptable salt thereof.
293. A medicament, wherein the medicament is I-47 or a pharmaceutically acceptable salt thereof.
294. The agent of any one of the preceding embodiments, wherein the agent specifically binds to CD38 as measured by SPR.
295. The agent of any one of the preceding embodiments, wherein the agent binds to CD38 with a Kd of no more than 200, 100, 50, 40, 30, 20, 10, or 5nM as measured by SPR.
296. The agent according to any one of the preceding embodiments, wherein the agent binds to CD38 as measured under the conditions described in the specification.
297. The agent according to any one of the preceding embodiments, wherein the agent binds to CD38 as measured under the conditions described in the examples.
298. The agent of any one of the preceding embodiments, wherein the agent recruits antibodies to target cells expressing CD 38.
299. The agent of any one of the preceding embodiments, wherein the agent provides a lesser reduction in non-diseased effector cells expressing CD38 as compared to the CD38 antibody.
300. A composition comprising an agent according to any one of the preceding embodiments and a population of cells.
301. The composition of embodiment 300, wherein the cell is a manipulated cell.
302. The composition of any one of embodiments 300 to 301, wherein the cell is a manufactured cell.
303. The composition of any one of embodiments 300-302, wherein the cells are suitable for cell-based therapy.
304. The composition of any one of embodiments 300 to 303, wherein the cell is or comprises an NK cell.
305. The composition of any one of embodiments 300 to 304, wherein the cell is or comprises an engineered NK cell.
306. The composition of any one of embodiments 300 to 305, wherein the cells are or comprise ex vivo expanded NK cells.
307. The composition of any one of embodiments 300 to 306, wherein the cells are or comprise memory-like NK cells.
308. The composition of any one of embodiments 300 to 307, wherein the cells are or comprise cytokine-induced memory-like NK cells.
309. The composition of any one of embodiments 300 to 303, wherein the cell is or comprises an NKT cell.
310. The composition of any one of embodiments 300 to 303, wherein the cell is or comprises a monocyte.
311. The composition of any one of embodiments 300 to 303, wherein the cell is or comprises a macrophage.
312. A pharmaceutical composition comprising an agent or composition according to any of the preceding embodiments and a pharmaceutically acceptable carrier.
313. The composition of any one of embodiments 300 to 312, wherein the composition comprises an immunoglobulin.
314. The composition of embodiment 313, wherein the immunoglobulin is or comprises an IgG.
315. The composition of any one of embodiments 313-314, wherein the immunoglobulin is an intravenous immunoglobulin.
316. The composition of any one of embodiments 313-315, wherein the immunoglobulin is an antibody against a particular antigen.
317. The composition of any of the preceding embodiments, wherein the composition is cryopreserved.
318. A method for treating a CD 38-associated condition, disorder or disease comprising administering to a subject suffering from the CD 38-associated condition, disorder or disease an effective amount of an agent or composition according to any of the preceding embodiments.
319. The method of embodiment 318, comprising administering to the subject a population of cells.
320. The method of embodiment 319, wherein the subject is subjected to both the agent or composition and the population of cells.
321. The method of any one of embodiments 319 to 320, wherein the cell is a manipulated cell.
322. The method of any one of embodiments 319 to 321, wherein the cell is a manufactured cell.
323. The method of any one of embodiments 319 to 322, wherein the cells are suitable for cell-based therapy.
324. The method of any one of embodiments 319 to 323, wherein the cell is an NK cell.
325. The method of any one of embodiments 319 to 324, wherein the cell is an engineered NK cell.
326. The method of any one of embodiments 319 to 325, wherein the cells are ex vivo expanded NK cells.
327. The method of any one of embodiments 319 to 326, wherein the cells are memory-like NK cells.
328. The method of any one of embodiments 319 to 327, wherein the cells are cytokine-induced memory-like NK cells.
329. The method according to any one of embodiments 319 to 323, wherein the cells are NKT cells.
330. The method of any one of embodiments 319 to 323, wherein the cell is a monocyte.
331. The method of any one of embodiments 319-323, wherein the cell is a macrophage.
332. The method of any one of embodiments 319-331, wherein the cell is administered concurrently with the agent or composition.
333. The method of any one of embodiments 319-332, wherein the cell and the agent or composition are administered simultaneously in the form of a composition comprising the cell and the agent or composition.
334. The method of any one of embodiments 319 to 331, wherein the cells are administered prior to administration of the agent or composition.
335. The method of any one of embodiments 319 to 331, wherein the cells are administered after administration of the agent or composition.
336. The method of any one of embodiments 318 to 335, wherein the method comprises administering an immunoglobulin.
337. The method of any one of embodiments 319 to 335, wherein the method comprises administering an immunoglobulin.
338. The method of any one of embodiments 336 to 337, wherein the immunoglobulin is or comprises IgG.
339. The method of any one of embodiments 336-338, wherein the immunoglobulin is an intravenous immunoglobulin.
340. The method of any one of embodiments 336 to 339, wherein the immunoglobulin is an antibody against a specific antigen.
341. The method of any one of embodiments 336-340, wherein the immunoglobulin is administered concurrently with the agent or composition.
342. The method of any one of embodiments 336-341, wherein the immunoglobulin and the agent or composition are administered simultaneously in the form of a composition comprising the immunoglobulin and the agent or composition.
343. The method of any one of embodiments 336-340, wherein the immunoglobulin is administered before or after the agent or composition.
344. The method of any one of embodiments 337-343, wherein the immunoglobulin is administered concurrently with the cells.
345. The method of any one of embodiments 336-339, wherein the immunoglobulin is administered simultaneously with the cell in a composition comprising the immunoglobulin and the cell.
346. The method of any one of embodiments 336-339, wherein the immunoglobulin is administered before or after the cell.
347. The method according to any of the preceding embodiments, wherein one or more doses of the cells according to any of the preceding embodiments and/or one or more doses of the cells according to any of the preceding embodiments and the agent according to any of the preceding embodiments are administered after administration of the agent.
348. The method of embodiment 347, wherein administration of the agent is a single dose of the agent.
349. The method of any one of embodiments 347 to 348, wherein one or more doses of the cells of any one of the preceding embodiments are administered after administration of the agent.
350. The method of any one of embodiments 347 to 349, wherein one or more doses of the cell according to any one of the preceding embodiments and the agent according to any one of the preceding embodiments are administered after administration of the agent.
351. The method of any one of embodiments 347-350, wherein for each dose of cells and agent, the cells are administered independently before, simultaneously with, or after the agent.
352. The method of any one of embodiments 347-350, wherein the cells and agent are administered in the same composition for at least one dose of the cells and agent.
353. A method, comprising:
a) providing a first compound comprising a target binding moiety as described in any one of the preceding embodiments and a first reactive group;
b) providing a second compound comprising an antibody binding moiety as described in any one of the preceding embodiments and a second reactive group; and
c) Reacting the first reactive group with the second reactive group to covalently link the target binding moiety to the antibody binding moiety.
354. The method of embodiment 353, wherein the reaction between the first reactive group and the second reactive group is or comprises an amidation reaction.
355. The method of embodiment 354, wherein one of the first reactive group and the second reactive group is or comprises
Figure BDA0003526174380002451
And the other is or comprises-NH2
356. The method of embodiment 353, wherein the reaction between the first reactive group and the second reactive group is a cycloaddition reaction.
357. The method of any one of embodiments 353-356, wherein the first compound is a compound of formula V or a salt thereof.
358. The method of any one of embodiments 353-357, wherein the first compound is a compound of formula V or a salt thereof, wherein
Figure BDA0003526174380002452
As described in any of the preceding embodiments
Figure BDA0003526174380002453
359. The method of any one of embodiments 353-358, wherein the second compound is a compound of formula IV, IV-a, IV-b, IV-c, or IV-d, or a salt thereof.
360. A method for manufacturing the agent or composition according to any one of the preceding embodiments, comprising reacting a first compound comprising the antibody-binding moiety and the alkyne with a second compound comprising the target-binding moiety and the azide, or reacting a first compound comprising the antibody-binding moiety and the azide with a second compound comprising the target-binding moiety and the alkyne.
361. The method of embodiment 360, wherein the alkyne is a ring strain activated alkyne.
362. The method of embodiment 360, wherein the alkyne is an alkyne within an 8-membered ring.
363. The method of embodiment 360, wherein the alkyne is an alkyne of a BCN moiety.
364. The process of any one of embodiments 360 to 363, wherein the reaction is carried out in the absence of a copper salt.
365. A method for recruiting an antibody to a target comprising or expressing CD38, comprising contacting the target with an agent or composition according to any of the preceding embodiments.
366. A method for recruiting immune activity to a target comprising or expressing CD38, comprising contacting the target with an agent according to any of the preceding embodiments.
367. The method of any one of embodiments 365-366, wherein the target is a tumor cell.
Example
As depicted in the examples below, in certain exemplary embodiments, the compounds are prepared according to the following general procedure. It is to be understood that while the general methods depict the synthesis of certain compounds of the present disclosure, the following general methods and other methods known to one of ordinary skill in the art may be applicable to all compounds as described herein and to subclasses and classes of each of these compounds.
Example 1. exemplary Synthesis of Compounds.
Exemplary preparation of I-1.
Figure BDA0003526174380002461
Figure BDA0003526174380002471
Peptide synthesis: the peptides were synthesized using standard Fmoc chemistry. An exemplary procedure is described below.
1) In N2DCM was added to a vessel containing the CTC resin (0.300mmol, 300mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (95.3mg, 0.240mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.30mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-PEG6-CH2CH2-COOH(2.00eq) HATU (1.9eq) and DIEA (4.0eq)
15 Fmoc-D-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
16 3-azidopropionic acid (3.00eq) HATU (2.85eq) and DIEA (6.00eq)
In one preparation:
the synthesis scale is as follows: 0.24 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification. Various schemes may be utilized. In one example:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 1.5 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, 200mL total)
Disulfide bond via I2MeOH formation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(35.0 mg).
A mixture of compound 3(5.0mg, 1.0eq) and compound 2(4.55mg, 1.0eq) was dissolved in DMF (0.5mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1482.3, 1112.2, 890.1, 741.8, etc.) were combined and lyophilized to give I-1 as a white solid (4.1mg, 42.9% yield, 95.4% purity). And (3) purification conditions:
Figure BDA0003526174380002491
Exemplary preparation of I-2.
Figure BDA0003526174380002501
Figure BDA0003526174380002511
Figure BDA0003526174380002521
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.300mmol, 300mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (95.3mg, 0.240mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.30mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380002522
Figure BDA0003526174380002531
The synthesis scale is as follows: 0.24 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 1.5 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, 200mL total)
Disulfide bond via I2MeOH formation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(35.0 mg).
A mixture of Compound 2(35.0mg, 1.0 equiv.) and DBCO-NHS (7.66mg, 1.1 equiv.) was dissolved in DMF (1.0mL) and DIEA (6.00 eq.) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (11.0mg, 32.4% yield).
Compound 4 was prepared by reacting the corresponding peptide dissolved in DMF (0.5mL) with 3-azidopropionic acid-NHS (0.97mg, 1.1 eq). DIEA (6.0eq) was then added slowly. The mixture was stirred at 20 ℃ for 2 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 4 was obtained as a white solid (5.50mg, 52.4% yield).
A mixture of compound 4(5.5mg, 1.0eq) and compound 3(5.10mg, 1.0eq) was dissolved in DMF (0.5mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1600.3, 1200.4, 960.5, etc.) were combined and lyophilized to give I-2 as a white solid (4.1mg, 38.7% yield, 95.7% purity). And (3) purification conditions:
Figure BDA0003526174380002541
exemplary preparation of I-3.
Figure BDA0003526174380002551
Figure BDA0003526174380002561
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.300mmol, 300mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (95.3mg, 0.240mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.30mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380002562
Figure BDA0003526174380002571
The synthesis scale is as follows: 0.24 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 1.5 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, 200mL total)
Disulfide bond via I2MeOH formation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(45.0 mg).
A mixture of Compound 2(45.0mg, 1.0eq) and BCN-NHS (6.48mg, 1.1eq) was dissolved in DMF (1.0mL) and DIEA (6.0eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (14.0mg, 28.6% yield).
A mixture of compound 3(14.0mg, 1.0eq) and compound 75(12.7mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1049.9, 840.0, 700.4, etc.) were combined and lyophilized to give I-3 as a white solid (11.1mg, 41.6% yield, 97.1% purity). And (3) purification conditions:
Figure BDA0003526174380002581
exemplary preparation of I-4.
Figure BDA0003526174380002582
Figure BDA0003526174380002591
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.300mmol, 300mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (95.3mg, 0.240mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.30mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-PEG8-CH2CH2-COOH(2.00eq) HATU (1.9eq) and DIEA (4.0eq)
15 Fmoc-D-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
16 Fmoc-D-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
The synthesis scale is as follows: 0.24mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 1.5 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H 2O (1: 1, 200mL total)
Disulfide bond via I2MeOH formation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(48.0 mg).
A mixture of Compound 2(48.0mg, 1.0eq) and BCN-NHS (9.37mg, 1.1eq) was dissolved in DMF (1.0mL) and DIEA (6.0eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (14.0mg, 28.6% yield).
A mixture of compound 3(14.0mg, 1.0eq) and compound 75(11.0mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1138.6, 911.2, 759.4, etc.) were combined and lyophilized to give I-4 as a white solid (15.9mg, 63.3% yield, 96.6% purity). And (3) purification conditions:
Figure BDA0003526174380002611
exemplary preparation of I-5.
Figure BDA0003526174380002612
Figure BDA0003526174380002621
Figure BDA0003526174380002631
Figure BDA0003526174380002641
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) In N2DCM was added to a vessel containing the CTC resin (0.300mmol, 300mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (95.3mg, 0.240mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.30mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-A1a-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-PEG3-CH2CH2-COOH(2.00eq) HATU (1.9eq) and DIEA (4.0eq)
15 Fmoc-D-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
The synthesis scale is as follows: 0.24mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 1.5 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, 200mL total)
Disulfide bond via I2MeOH formation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O,B: ACN) to give compound 2(42.0 mg).
A mixture of Compound 2(42.0mg, 1.0eq) and BCN-NHS (7.11mg, 1.1eq) was dissolved in DMF (1.0mL) and DIEA (6.0eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (15.0mg, 32.7% yield).
A mixture of compound 4(13.5mg, 1.0eq) and compound 3(15.0mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 983.8, 787.6, etc.) were combined and lyophilized to give I-5 as a white solid (10.5mg, 36.8% yield, 95.1% purity). And (3) purification conditions:
Figure BDA0003526174380002661
exemplary preparation of I-6.
Figure BDA0003526174380002662
Figure BDA0003526174380002671
Figure BDA0003526174380002681
Compound 1(2.00g, 4.37mmol), Fmoc-NH-PEG3-CH2CH2N3A mixture of (1.00g, 4.59mmol), EDCI (1.68g, 8.74mmol), HOBt (1.18g, 8.74mmol) was dissolved in DCM (20.0mL) and the reaction was stirred at 15 ℃ for 16 h. The solution was then diluted with DCM (100mL) and 1M HCl (30mL), H2O (30mL), brine (30mL), over anhydrous Na2SO4Drying and concentration under reduced pressure gave compound 2(3.00g, crude material) as a colorless oil.
With TFA/H at 15 deg.C2Compound 2(3.00g, crude material) was treated with O (95/5, ca. 20mL) for 1 hour. The solvent was then removed under reduced pressure and chromatographed by flash C18 (C)
Figure BDA0003526174380002682
120g
Figure BDA0003526174380002683
C18 flash column, eluent: 0 to 100% MeCN/H2O, 75mL/min) to give compound 3 as a white solid (1.50g, 2.24mmol, 49.2% yield).
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) DCM (10.0mL) was added to a vessel containing the CTC resin (2.0mmol, 2.0g, 1.0mmol/g) and Fmoc-Asp (OtBu) -OH (658mg, 1.60mmol, 0.80eq) with N2 bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (2.0mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation buffer was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Step (ii) of Material Coupling reagent
1 Fmoc-Asp(OtBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-HomoNle-OH(2.00eq) HATU (1.90eq) and DIEA (4.00eq)
3 Fmoc-Bip-OH(2.00eq) HATU (1.90eq) and DIEA (4.00eq)
4 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Asp(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
9 Fmoc-Tyr(tBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-HomoNle-OH(2.00eq) HATU (1.90eq) and DIEA (4.00eq)
11 Fmoc-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Compound 3(2.00eq) HATU (1.90eq) and DIEA (4.00eq)
The synthesis scale is as follows: 1.6mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After coupling, the Fmoc group was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to the flask containing the side chain protected peptide was added the lysis solution (20% HFIP/DCM, 80mL) at room temperature. Cracking in continuous N2Bubbling was performed twice (30 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 4(2.9g, crude material) as a white solid.
A solution of compound 4(2.9g, 1.0eq), HOBt (308mg, 2.0eq), TBTU (732mg, 2.0eq), DIEA (0.85mL, 4.0eq) in DMF (800mL) was stirred at 15 ℃ for 1 hour. When cyclization was complete, the solution was then diluted with EA (1.5L), washed with 1M HCl (600mL), brine (400 mL. times.4), and dried over anhydrous Na2SO4Drying and concentration under reduced pressure gave compound 5(3.2g, crude material) as a colorless oil.
The resulting mixture was stirred continuously at 15 ℃ for 1.5 hours with TFA/TIS/H2Deprotection of the crude cyclic peptide was performed by O (95/2.5/2.5, 40mL total) treatment of Compound 5(3.2g, crude material). The solution was wet milled with cold isopropyl ether (500mL) and the precipitate was collected by centrifugation (3 minutes at 3000 rpm). The precipitate (deprotected peptide) was washed twice with isopropyl ether (50 mL each) and then dried in vacuo for 2 h. The residue was purified by preparative HPLC (acidic conditions, TFA) to give compound 6(600 mg).
Another example is:
1) in N2DCM was added to a vessel containing the CTC resin (0.500mmol, 500mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (159mg, 0.400mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-PEG3-CH2CH2COOH(2.00eq) HATU (1.90eq) and DIEA (4.0eq)
15 Fmoc-D-Lys(Dde)-OH(2.00eq) HATU (1.90eq) and DIEA (4.0eq)
16 Fmoc-PEG3-CH2CH2COOH(2.00eq) HATU (1.90eq) and DIEA (4.0eq)
17 Boc-D-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
18 Palmitic acid (3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
The synthesis scale is as follows: 0.4mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After cycle 17, 3% NH2NH2H2O/DMF was used to remove Dde and then to couple palmitic acid to the side chain of D-Lys.
After the last coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O(1∶1,Total 600mL) in
Disulfide bond via I2MeOH was formed and stirred for 20 minutes, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 8(55.0 mg).
A mixture of compound 8(55.0mg, 1.0eq) and BCN-NHS (7.20mg, 1.1eq) was dissolved in DMF (1mL) and DIEA (6.0eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 9 was obtained as a white solid (19.0mg, 32.0% yield).
A mixture of compound 9(19.0mg, 1.0eq) and compound 6(13.5mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1126.0, 901.2, 751.1, etc.) were combined and lyophilized to give I-6 as a white solid (12.7mg, 39.2% yield, 92.4% purity). And (3) purification conditions:
Figure BDA0003526174380002711
exemplary preparation of I-7.
Figure BDA0003526174380002721
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.500mmol, 500mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (159mg, 0.400mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-Gly-Gly-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
15 Fmoc-Lys(Dde)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
16 Fmoc-Glu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
17 Fmoc-Ser(tBu)-Ser[psiMe,-Mepro]-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
18 Fmoc-Gly-Gly-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
19 Boc-D-Arg(Pbf)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
20 Palmitic acid (3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
The synthesis scale is as follows: 0.4mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the 19 th cycle, 3% NH2NH2H2O/DMF was used to remove Dde and then to couple palmitic acid to the side chain of Lys.
After the last coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 600mL)
Disulfide bond via I2MeOH was formed and stirred for 20 minutes, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(63.0 mg).
A mixture of Compound 2(63.0mg, 1.0eq) and BCN-NHS (7.47mg, 1.1eq) was dissolved in DMF (1mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (20.0mg, 29.8% yield).
A mixture of compound 3(20.0mg, 1.0eq) and compound 75(13.9mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1219.3, 975.4, etc.) were combined and lyophilized to give I-7 as a white solid (11.7mg, 34.5% yield, 96.8% purity). And (3) purification conditions:
Figure BDA0003526174380002741
Exemplary preparation of I-8.
Figure BDA0003526174380002751
Figure BDA0003526174380002761
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.500mmol, 500mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (159mg, 0.400mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380002762
Figure BDA0003526174380002771
The synthesis scale is as follows: 0.4mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
Pd (PPh)3)4And phenylsilanes for De-OAll.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage cocktail (95% TFA/2.5% EDT/2.5% H) at room temperature 2O/2.5% TIS) and stirred for 2 hours.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 600mL)
By NaHCO3The pH was adjusted to 8 and stirred for 16 hours and disulfide bonds were formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(75.0 mg).
A mixture of Compound 2(75.0mg, 1.0eq) and BCN-NHS (8.91mg, 1.1eq) was dissolved in DMF (2mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (35.0mg, 43.8% yield).
A mixture of compound 3(35.0mg, 1.0eq) and compound 75(24.4mg, 1.0eq) was dissolved in DMF (2.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1218.3, 974.8, 812.2, etc.) were combined and lyophilized to give I-8 as a white solid (29.3mg, 49.7% yield, 97.6% purity). And (3) purification conditions:
Figure BDA0003526174380002781
Exemplary preparation of I-9.
Figure BDA0003526174380002782
Figure BDA0003526174380002791
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.500mmol, 500mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (159mg, 0.400mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-Gly-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
15 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
16 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
17 Fmoc-D-Glu(OAll)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
18 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
19 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
20 Fmoc-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
21 Fmoc-piperidine-4-carboxylic acid (3.00eq) HATU (2.85eq) and DIEA (6.00eq)
22 12-Aminododecanoic acid tert-butyl ester (3.00eq) HOAt (3.0eq) and DIC (3.00eq)
The synthesis scale is as follows: 0.4mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
Pd (PPh)3)4And phenylsilane was used to remove the aly group on the side chain of D-Glu.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage cocktail (95% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 600mL)
By NaHCO3The pH was adjusted to 8 and stirred for 16 hours, and the first disulfide bond was formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(42.0 mg).
A mixture of Compound 2(42.0mg, 1.0eq) and BCN-NHS (5.06mg, 1.1eq) was dissolved in DMF (1mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (13.0mg, 29.0% yield).
A mixture of compound 3(13.0mg, 1.0eq) and compound 75(9.18mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1207.9, 966.8, 805.6, etc.) were combined and lyophilized to give I-9 as a white solid (10.9mg, 49.1% yield, 97.1% purity). And (3) purification conditions:
Figure BDA0003526174380002811
exemplary preparation of I-10.
Figure BDA0003526174380002821
Azide-containing compound 75(1.0eq) and the corresponding alkyne-containing compound 3(1.0eq), prepared using similar techniques described above, were dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1019.0, 849.0, etc.) were combined and lyophilized to give I-10 as a white solid (7.3mg, 33.2% yield, 98.9% purity). And (3) purification conditions:
Figure BDA0003526174380002822
Exemplary preparation of I-11.
Figure BDA0003526174380002831
Figure BDA0003526174380002841
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM (2.00mL) was added to the CTC-containing resin (0.50mmol, 0.45g, 1.10mmol/g) and Fmoc-NH-PEG with bubbling6-CH2CH2COOH (0.30g, 0.50mmol, 1.00 eq).
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation buffer was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Step (ii) of Material Coupling reagent
1 Fmoc-NH-PEG6-CH2CH2COOH(1.00eq) DIEA(4.00eq)
2 Fmoc-Arg(Pbf)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Arg(Pbf)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Inp-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Boc2O(3.00eq) DIEA(6.00eq)
The synthesis scale is as follows: 0.50mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min. The coupling reaction was monitored by ninhydrin test and the resin was washed 5 times with DMF.
After the last amino acid coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage:
to the flask containing the side chain protected peptide was added lysis solution (20% HFIP/DCM, 50mL) at room temperature. Cracking in continuous N 2Bubbling was performed twice (30 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 1(0.50g, crude material) as a white solid.
A solution of Compound 1(50.0mg, 36.0. mu. mol), N- [3- (3-aminopropylcarbamoyl) -4-methoxy-phenyl ] imidazo [2, 1-b ] thiazole-6-carboxamide (17.0mg, 36.3. mu. mol, TFA salt), EDCI (69.1mg, 362. mu. mol) in pyridine (0.50mL) was stirred at 25 ℃ for 16 h. Pyridine was then removed under reduced pressure to give a residue, to which TFA (1.00mL) was then added and stirred at 25 ℃ for 1 hour to remove the Boc protecting group. The solvent was removed under reduced pressure and the residue was purified by preparative HPLC (acidic conditions, TFA) to give compound 2(TFA salt, 35.0mg, 30.9 μmol, 85.4% yield) as a white solid.
A solution of Compound 2(35.0mg, 28.1. mu. mol, TFA salt), [ (1R, 8S) -9-bicyclo [6.1.0] non-4-ynyl ] methyl carbonate (2, 5-dioxopyrrolidin-1-yl) ester (12.2mg, 42.1. mu. mol), DIEA (36.3mg, 281. mu. mol, 49. mu.L) in DMF (0.20mL) was stirred at 25 ℃ for 3 hours. The solution was then purified by preparative HPLC (acidic conditions, TFA) to give compound 3(30.0mg, 22.9 μmol, 81.6% yield) as a white solid.
A solution of compound 75(39.7mg, 19.8. mu. mol), compound 3(26.0mg, 19.8. mu. mol), DIEA (10.2mg, 79.4. mu. mol, 14. mu.L) in DMF (0.70mL) was stirred at 25 ℃ for 3 hours. When the click reaction was complete (indicated by LCMS, MS observations: m/z 1102.3, 826.8, 661.6, etc.), the solution was purified directly by preparative HPLC (acidic conditions, TFA) to afford Compound I-11(29.0mg, 8.33. mu. mol, 41.9% yield, 98.4% purity) as a white solid after lyophilization. And (3) purification conditions:
Figure BDA0003526174380002851
Figure BDA0003526174380002861
exemplary preparation of I-12.
Figure BDA0003526174380002862
Figure BDA0003526174380002871
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM (2.00mL) was added to the CTC-containing resin (0.50mmol, 0.45g, 1.1mmol/g) and Fmoc-NH-PEG with bubbling6-CH2CH2COOH (0.30g, 0.50mmol, 1.00 eq).
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation buffer was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380002872
Figure BDA0003526174380002881
The synthesis scale is as follows: 0.50 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min. The coupling reaction was monitored by ninhydrin test and the resin was washed 5 times with DMF.
After the last amino acid coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage:
to the flask containing the side chain protected peptide was added lysis solution (20% HFIP/DCM, 50mL) at room temperature. Cracking in continuous N2Bubbling was performed twice (30 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 1(0.50g, crude material) as a white solid.
A solution of compound 1(50.0mg, 36.1. mu. mol), 1- (3-aminopropyl) -N- [5- (2-furyl) -1, 3, 4-thiadiazol-2-yl ] -2-oxo-3H-benzimidazole-5-carboxamide (18.0mg, 36.1. mu. mol, TFA salt), EDCI (69.3mg, 362. mu. mol) in pyridine (0.50mL) was stirred at 25 ℃ for 16H. Pyridine was then removed under reduced pressure. To the residue was added TFA (1mL) and stirred at 25 ℃ for 1 hour. The solvent was removed under reduced pressure. The residue was purified by preparative HPLC (acidic conditions, TFA) to give compound 2(TFA salt, 25.0mg, 21.8 μmol, 60.4% yield) as a white solid.
A solution of Compound 2(25.0mg, 19.8. mu. mol, TFA salt), [ (1R, 8S) -9-bicyclo [6.1.0] non-4-ynyl ] methyl carbonate (2, 5-dioxopyrrolidin-1-yl) ester (5.8mg, 19.8. mu. mol), DIEA (12.8mg, 99.4. mu. mol, 18. mu.L) in DMF (0.2mL) was stirred at 25 ℃ for 3 hours. The solution was purified by preparative HPLC (acidic conditions, TFA) to give compound 3(15mg, 11.3 μmol, 57.1% yield) as a white solid.
A solution of compound 75(22.7mg, 11. mu. mol), compound 3(15.0mg, 11. mu. mol) in DMF (0.7mL) was stirred at 25 ℃ for 3 hours. The solution was purified by preparative HPLC (acidic conditions, TFA) to give I-12(25.9mg, 7.65 μmol, 67.3% yield, 98.5% purity) as a white solid. LCMS MS observations: m/z 1105.8, 829.5, 663.7 and the like. And (3) purification conditions:
Figure BDA0003526174380002882
Figure BDA0003526174380002891
exemplary preparation of I-13.
Figure BDA0003526174380002892
Figure BDA0003526174380002901
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1. in N2DCM (2.00mL) was added to the CTC-containing resin (0.50mmol, 0.45g, 1.1mmol/g) and Fmoc-NH-PEG with bubbling6-CH2CH2COOH (0.30g, 0.50mmol, 1.00 eq).
2. DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3. MeOH (0.50mL) was added and mixed for 30 minutes.
4. Drained and washed 5 times with DMF.
5. 20% piperidine/DMF was added and the reaction was for 30 min.
6. Drained and washed 5 times with DMF.
7. Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation buffer was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8. Steps 4 to 7 are repeated for the next amino acid coupling.
Step (ii) of Material Coupling reagent
1 Fmoc-NH-PEG6-CH2CH2COOH(1.00eq) DIEA(4.00eq)
2 Fmoc-Arg(Pbf)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Arg(Pbf)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Inp-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Boc2O(3.00eq) DIEA(6.00eq)
The synthesis scale is as follows: 0.50mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min. The coupling reaction was monitored by ninhydrin test and the resin was washed 5 times with DMF.
After the last amino acid coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage:
to the flask containing the side chain protected peptide was added lysis solution (20% HFIP/DCM, 50mL) at room temperature. Cracking in continuous N2Bubbling was performed twice (30 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 1(0.50g, crude material) as a white solid.
A solution of Compound 1(50.0mg, 36.1. mu. mol), 1- (3-aminopropyl) -2-methyl-N- [4- (2-pyridyl) thiazol-2-yl ] benzimidazole-5-carboxamide (14.2mg, 36.1. mu. mol, TFA salt), EDCI (69.3mg, 362. mu. mol) in pyridine (0.50mL) was stirred at 25 ℃ for 16 h. Pyridine was then removed under reduced pressure. To the residue was added TFA (1.00mL) and stirred at 25 ℃ for 1 hour to remove the Boc group. After deprotection, the solvent was removed under reduced pressure and the residue was purified by preparative HPLC (acidic conditions, TFA) to give compound 2(29.0mg, 16.5 μmol, 45.6% yield) as a white solid.
A solution of Compound 2(29.0mg, 22.9. mu. mol, TFA salt), [ (1R, 8S) -9-bicyclo [6.1.0] non-4-ynyl ] methyl carbonate (2, 5-dioxopyrrolidin-1-yl) ester (6.7mg, 22.9. mu. mol), DIEA (14.8mg, 114. mu. mol, 14.8. mu.L) in DMF (0.20mL) was stirred at 25 ℃ for 3 hours. The solution was purified by preparative HPLC (acidic conditions, TFA) to give I-13(TFA salt, 20.0mg, 15.0 μmol, 65.7% yield) as a white solid.
A solution of compound 75(30.2mg, 15.1. mu. mol), compound 3(20.0mg, 15.1. mu. mol) in DMF (0.70mL) was stirred at 25 ℃ for 3 hours. The solution was directly purified by preparative HPLC (acidic conditions, TFA) to afford I-13(27.2mg, 8.06 μmol, 53.1% yield, 99.0% purity) as a white solid after lyophilization. LCMS MS observations: 1108.6, 831.4, 665.4, etc. And (3) purification conditions:
Figure BDA0003526174380002911
exemplary preparation of I-14.
Figure BDA0003526174380002921
Figure BDA0003526174380002931
Compound 1(1.00g, 2.4mmol), Fmoc-NH-PEG3-CH2CH2N3A mixture of (1.10g, 2.4mmol), EDCI (688mg, 3.6mmol), HOBt (486mg, 3.6mmol) was dissolved in DCM (30.0mL) and the reaction was stirred at 15 ℃ for 16 h. The solution was then diluted with DCM (100mL) and 1M HCl (30mL), H2O (30mL), brine (30mL), over anhydrous Na 2SO4Drying and concentration under reduced pressure gave compound 2(3.00g, crude material) as a colorless oil.
With TFA/H at 15 deg.C2Compound 2(3.00g, crude material) was treated with O (95/5, ca. 20mL) for 1 hour. The solvent was then removed under reduced pressure. The solution was then diluted with DCM (100mL) and 1M HCl (30mL), H2O (30mL), brine (30mL), over anhydrous Na2SO4Drying, and concentrating under reduced pressure to obtain crude material. The crude material (2.6g, 3.67mmol, 1.0eq) was first dissolved in DCM and DIEA (1.92mL, 3eq) was then added and mixed well, finally Boc was added dropwise at 25 ℃2Solution O (1.20g, 5.51mmol, 1.27mL, 1.5eq, in 20mL DCM). The reaction mass is brought to 25 DEG CStirred for 3 hours and then chromatographed by flash C18 (
Figure BDA0003526174380002942
120g
Figure BDA0003526174380002943
C18 flash column, eluent: 0 to 100% MeCN/H2O, 75mL/min) to give compound 3 as a white solid (1.46g, 2.24mmol, 49.2% yield).
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.200mmol, 200mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (63.5mg, 0.16mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.20mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380002941
Figure BDA0003526174380002951
The synthesis scale is as follows: 0.16mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H2O/2.5% TIS) at room temperature and stirred for 1.5H.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, 100mL total)
By NaHCO3The pH was adjusted to 8 and stirred for 8 hours and disulfide bonds were formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 5(30.0 mg).
Another example is:
1) in N2DCM was added to a vessel containing the CTC resin (0.500mmol, 500mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (130mg, 0.40mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Step (ii) of Material Coupling reagent
1 Fmoc-Abu-OH(0.8eq) DIEA(4.0eq)
2 Fmoc-heptanoic acid (2.0eq) HATU (1.9eq) and DIEA (4.0eq)
3 Fmoc-Bip-OH(2.0eq) HATU (1.9eq) and DIEA (4.0eq)
4 Fmoc-Leu-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
5 Fmoc-Val-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
6 Fmoc-Gly-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
7 Fmoc-Asp(tBu)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
8 Fmoc-His(Trt)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
9 Fmoc-Tyr(tBu)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
10 Fmoc-heptanoic acid (2.0eq) HATU (1.9eq) and DIEA (4.0eq)
11 Fmoc-Arg(Pbf)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
12 Fmoc-Ala-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
13 Compound 3(2.0eq) HATU (1.9eq) and DIEA (4.0eq)
The synthesis scale is as follows: 0.4 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to the flask containing the side chain protected peptide was added the lysis solution (20% HFIP/DCM, 20mL) at room temperature. Cracking in continuous N2Bubbling was performed twice (30 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 6(1.0g, crude material) as a white solid.
A solution of compound 6(1.0g, 1.0eq), HOBt (101mg, 2.0eq), TBTU (240mg, 2.0eq), DIEA (0.85mL, 4.0eq) in DMF (400mL) was stirred at 15 ℃ for 1 hour. When cyclization was complete, the solution was then diluted with EA (1.5L), washed with 1M HCl (600mL), brine (400 mL. times.4), and dried over anhydrous Na2SO4Dried and concentrated under reduced pressure to give the crude material (1.1g, crude material) as a colorless oil.
The resulting mixture was stirred continuously at 15 ℃ for 1.5 hours with TFA/H2Deprotection of the crude cyclic peptide was performed by O (97.5/2.5, 20mL total) treating the cyclized peptide (1.1g, crude). The solution was wet milled with cold isopropyl ether (250mL) and the precipitate was collected by centrifugation (3 minutes at 3000 rpm). The precipitate (deprotected peptide) was washed twice with isopropyl ether (50 mL each) and then dried in vacuo for 2 h. The residue was purified by preparative HPLC (acidic conditions, TFA) to give compound 7(150 mg).
A mixture of compound 7(150mg, 1.0eq) and 2- (4- (6-methyl-1, 2, 4, 5-tetrazin-3-yl) phenyl) acetic acid 2, 5-dioxopyrrolidin-1-yl ester (27.8mg, 1.1eq) was dissolved in DMF (4.0mL) and then DIEA (6.0eq) was slowly added. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 8 was obtained as a white solid (48mg, 28.9% yield).
A mixture of Compound 8(11.0mg, 1.0eq) and Compound 5(9.94mg, 1.0eq) was dissolved in DMF/H2O (4: 1, 1.5mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1358.2, 1018.7, 815.4, etc.) were combined and lyophilized, yielding a white solidI-14 as (2.2mg, 10.5% yield, 76.4% purity). And (3) purification conditions:
Figure BDA0003526174380002971
exemplary preparation of I-15.
Figure BDA0003526174380002972
Figure BDA0003526174380002981
Figure BDA0003526174380002991
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) DMF was added to a resin containing Rink amide MBHA (1.00mmol, 2.27g, substrate: 0.440mmol/g) and allowed to expand for 2 hours.
2) Drained and washed 3 times with DMF.
3) Add 20% piperidine/DMF and mix for 30 min.
4) Drained and washed 3 times with DMF.
5) Fmoc-amino acid solution was added and mixed for 30 seconds, then coupling reagent was added, and the reaction was continued N2The coupling reaction was continued for 1 hour with bubbling.
6) Repeat steps 2 through 5 for the next amino acid coupling.
Figure BDA0003526174380002992
Figure BDA0003526174380003001
The synthesis scale is as follows: 1.0mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage cocktail (95% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 600mL)
By NaHCO3The pH was adjusted to 8 and stirred for 16 hours, and the first disulfide bond was formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(218mg, 7.46% yield).
Dissolve Compound 2(218mg) in ACN/H2O (50.0mL), and then 1M HCl was added to adjust the pH to 1, then 0.1M I was added dropwise2AcOH until the mixture turns brown. The mixture was then stirred at 20 ℃ for 10 hours. LCMS showed reaction completion. The mixture was purified by preparative HPLC (TFA conditions) to give compound 3(100mg, 48.2% yield) as a white solid.
A mixture of compound 3(100mg, 1.0eq) and BCN-NHS (11.5mg, 1.1eq) was dissolved in DMF (3mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 30 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 4 was obtained as a white solid (29.0mg, 27.4% yield).
A mixture of compound 4(29.0mg, 1.0eq) and compound 75(19.6mg, 1.1eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 15 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1238.0, 990.2, etc.) were combined and lyophilized to give I-15 as a white solid (30.6mg, 59.7% yield, 97.6% purity). And (3) purification conditions:
Figure BDA0003526174380003011
Exemplary preparation of I-16.
Figure BDA0003526174380003012
Figure BDA0003526174380003021
Figure BDA0003526174380003031
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) DMF was added to a resin containing Rink amide MBHA (1.00mmol, 2.27g, substrate: 0.440mmol/g) and allowed to expand for 2 hours.
2) Drained and washed 3 times with DMF.
3) Add 20% piperidine/DMF and mix for 30 min.
4) Drained and washed 3 times with DMF.
5) Fmoc-amino acid solution was added and mixed for 30 seconds, then coupling reagent was added, and the reaction was continued N2The coupling reaction was continued for 1 hour with bubbling.
6) Repeat steps 2 through 5 for the next amino acid coupling.
Figure BDA0003526174380003032
Figure BDA0003526174380003041
The synthesis scale is as follows: 1.0 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage cocktail (95% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 600mL)
By NaHCO3The pH was adjusted to 8 and stirred for 16 hours, and the first disulfide bond was formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA)2O, B: ACN) to give compound 2(248mg, 8.84% yield).
Compound 2(248mg) was dissolved in ACN/H2O (50.0mL), and then 1M HCl was added to adjust the pH to 1, then 0.1M I was added dropwise2AcOH until the mixture turns brown. The mixture was then stirred at 20 ℃ for 10 hours. LCMS showed reaction completion. Tong (Chinese character of 'tong')The mixture was purified by preparative HPLC (TFA conditions) to give compound 3(110mg, 44.4% yield) as a white solid.
A mixture of compound 3(110mg, 1.0eq) and BCN-NHS (13.2mg, 1.1eq) was dissolved in DMF (3mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 30 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 4 was obtained as a white solid (21.0mg, 17.9% yield).
A mixture of compound 4(21.0mg, 1.0eq) and compound 75(14.8mg, 1.1eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 15 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1209.4, 967.7, etc.) were combined and lyophilized to give I-16 as a white solid (20.4mg, 60.0% yield, 99.4% purity). And (3) purification conditions:
Figure BDA0003526174380003051
Exemplary preparation of I-17.
Figure BDA0003526174380003052
Figure BDA0003526174380003061
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (1.00mmol, 1.00g, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (3218mg, 0.800mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (1.00mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-Gly-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
15 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.0eq)
1 6 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
17 Fmoc-D-Glu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
18 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
19 Fmoc-D-gamaGlu(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
20 Fmoc-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
21 Fmoc-piperidine-4-carboxylic acid (3.00eq) HATU (2.85eq) and DIEA (6.00eq)
The synthesis scale is as follows: 0.8mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage cocktail (95% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 800mL)
By NaHCO3The pH was adjusted to 8 and stirred for 16 hours and disulfide bonds were formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
The crude peptide was purified by preparative HPLC (TFA conditions) to give compound 2(102 mg).
A mixture of compound 2(102mg, 1.0eq) and BCN-NHS (13.3mg, 1.1eq) was dissolved in DMF (2mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (43.0mg, 39.4% yield).
A mixture of compound 3(43.0mg, 1.0eq) and compound 75(32.6mg, 1.0eq) was dissolved in DMF (2.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1158.8, 927.2, 772.7, etc.) were combined and lyophilized to give I-17 as a white solid (41.1mg, 56.9% yield, 98.4% purity). And (3) purification conditions:
Figure BDA0003526174380003081
exemplary preparation of I-18.
Figure BDA0003526174380003091
Figure BDA0003526174380003101
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.300mmol, 300mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (95.3mg, 0.240mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.30mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380003102
Figure BDA0003526174380003111
The synthesis scale is as follows: 0.24 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last coupling, the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage mixture (92.5% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 1.5 hours.
After filtration, cold isopropyl ether was added to the solution and the peptide was collected by centrifugation (3 minutes at 3000 rpm) precipitation.
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, 200mL total)
Disulfide bond via I2MeOH formation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
By preparative HPLC (A: H with 0.075% TFA) 2O, B: ACN) to give compound 2(15.0 mg).
A mixture of compound 3(6.0mg, 1.0eq) and compound 2(4.93mg, 1.0eq) was dissolved in DMF (0.5mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1170.4, 939.4, 783.0, etc.) were combined and lyophilized to give I-18 as a white solid (7.1mg, 64.9% yield, 96.1% purity). And (3) purification conditions:
Figure BDA0003526174380003112
Figure BDA0003526174380003121
exemplary preparation of I-19.
Figure BDA0003526174380003122
Figure BDA0003526174380003131
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (0.500mmol, 500mg, 1.00mmol/g) and Fmoc-Thr (tBu) -OH (159mg, 0.400mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.50mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-PEG8-CH2CH2COOH(2.00eq) HATU (1.9eq) and DIEA (4.0eq)
15 FMOC-O- (phenylmethylphosphato) -serine (3.00eq) HATU (2.85eq) and DIEA (6.0eq)
16 FMOC-O- (phenylmethylphosphato) -serine (6.00eq) HATU (5.70eq) and DIEA (12.00eq)
20 Fmoc-Gly-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
21 Fmoc-piperidine-4-carboxylic acid (3.00eq) HATU (2.85eq) and DIEA (6.00eq)
The synthesis scale is as follows: 0.4 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to a flask containing the side chain protected peptide was added the cleavage cocktail (95% TFA/2.5% EDT/2.5% H) at room temperature2O/2.5% TIS) and stirred for 2 hours.
The peptide was precipitated with cold isopropyl ether and collected by centrifugation (3 minutes at 3000 rpm).
The precipitate was washed twice more with cold isopropyl ether.
The crude peptide was dried in vacuo for 2 hours.
Dissolving the crude peptide in ACN/H2O (1: 1, total 600mL)
By NaHCO3The pH was adjusted to 8 and stirred for 16 hours, and the first disulfide bond was formed via air oxidation, with completion of the reaction indicated by LCMS.
The reaction mixture was lyophilized to give the crude peptide.
The crude peptide was purified by preparative HPLC (TFA conditions) to give compound 2(80.0 mg).
A mixture of Compound 2(80.0mg, 1.0eq) and BCN-NHS (10.8mg, 1.1eq) was dissolved in DMF (1mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 20 ℃ for 8 hours. LCMS showed reaction completion. The mixture was then directly purified by preparative HPLC (TFA conditions) and compound 3 was obtained as a white solid (27.0mg, 31.4% yield).
A mixture of compound 3(27.0mg, 1.0eq) and compound 75(21.2mg, 1.0eq) was dissolved in DMF (1.0mL) and the reaction was stirred at 20 ℃ for 8 h. LCMS showed the reaction was complete, and then the mixture was directly purified by preparative HPLC. Fractions with the desired m/z (e.g., 1136.3, 909.4, 757.9, etc.) were combined and lyophilized to give I-19 as a white solid (28.8mg, 59.7% yield, 90.4% purity). And (3) purification conditions:
Figure BDA0003526174380003151
Exemplary preparation of I-24.
Figure BDA0003526174380003161
Figure BDA0003526174380003171
Figure BDA0003526174380003181
Figure BDA0003526174380003191
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) in N2DCM was added to a vessel containing the CTC resin (2.00mmol, 2.00g, 1.00mmol/g) and Fmoc-Cys (Trt) -OH (938mg, 1.60mmol, 0.80eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (0.10mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) Steps 4 to 7 are repeated for the next amino acid coupling.
Figure BDA0003526174380003192
Figure BDA0003526174380003201
The synthesis scale is as follows: 1.6mmol
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to the flask containing the side chain protected peptide was added the lysis solution (20% HFIP/DCM) at room temperature. Cracking in continuous N2Bubbling was performed twice (20 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 1(3.1g, crude material) as a white solid.
Another example is:
1) in N2DCM was added to a vessel containing CTC resin (2.00mmol, 2.00g, 1.00mmol/g) and hexane-1, 6-diamine (256mg, 2.20mmol, 1.10eq) with bubbling.
2) DIEA (4.00eq) was added dropwise and mixed for 2 hours.
3) MeOH (2.00mL) was added and mixed for 30 minutes.
4) Drained and washed 5 times with DMF.
5) 20% piperidine/DMF was added and the reaction was for 30 min.
6) Drained and washed 5 times with DMF.
7) Fmoc-amino acid solution was added first and mixed for 30 seconds, then activation solution was added, and the reaction was continued for N2The coupling reaction was continued for 1 hour with bubbling.
8) The above steps 4 to 7 were repeated for the coupling of the following amino acids.
Figure BDA0003526174380003202
Figure BDA0003526174380003211
The synthesis scale is as follows: 2.0 mmol.
DMF with 20% piperidine was used for Fmoc deprotection for 30 min.
The coupling reaction was monitored by ninhydrin test.
After the last amino acid coupling, the N-terminal Fmoc was removed and the resin was washed 3 times with MeOH and then dried in vacuo.
Peptide cleavage and purification:
to the flask containing the side chain protected peptide was added the lysis solution (20% HFIP/DCM) at room temperature. Cracking in continuous N 2Bubbling was performed twice (20 minutes each).
After filtration, the filtrate was concentrated under reduced pressure, and the residue was dried in a lyophilizer to give compound 2(800mg, crude material) as a white solid.
First a mixture of compound 2(500mg, 1.0eq) and compound 1(1.35g, 1.2eq) was dissolved in DMF (5.0mL), followed by the addition of HOAt (2.0eq, pre-dissolved in DMF) and DIC (2.0 eq). The mixture was stirred at 30 ℃ for 8 hours until LCMS appearedIndicating that the coupling is complete. The mixture was concentrated under reduced pressure to remove the solvent. To the crude material was added 30mL of deprotected mixture (95% TFA/2.5% TIS/2.5% H)2O/2.5% EDT) to remove all protecting groups, and this reaction was continued at room temperature (15 to 25 ℃) for 2 hours with continuous stirring. The peptide was then precipitated with cold tert-butyl methyl ether (150mL) and centrifuged (3 min at 3000 rpm). The precipitate was collected and washed two more times with cold tert-butyl methyl ether (150mL each) and the resulting crude peptide was then dried in vacuo for 2 hours. The deprotected peptide exposes two Cys, which are designed to form a first disulfide bond by free oxidation. Thus, the crude peptide is then dissolved in ACN/H2To O (300mL), 1M NaHCO was added3Until the pH reached 8 and finally the resulting solution was stirred with make-up air for 16 hours. LCMS indicated formation of the first disulfide bond, and then the solution was lyophilized to dryness. The residue was directly purified by preparative HPLC (TFA conditions) to give compound 5(150mg, 13.7% yield) as a white solid.
Dissolve Compound 5(150mg) in ACN/H2O (50.0mL), and then 1M HCl was added to adjust the pH to 1, then 0.1M I was added dropwise2AcOH until the mixture turns brown. The mixture was then stirred at 20 ℃ for 10 hours. LCMS showed reaction completion. The mixture was purified by preparative HPLC (TFA conditions) to give compound 6(70mg, 49.0% yield) as a white solid.
Compound 6(70.0mg, 1.0eq) and BCN-NHS (7.50mg, 1.1eq) were dissolved in DMF (3.00mL) and DIEA (6.00eq) was added slowly. The mixture was stirred at 30 ℃ for 8 hours. LCMS indicated the coupling reaction was complete. In addition, DMF with 20% piperidine was added for Fmoc removal (30 min). The mixture was directly purified by preparative HPLC, and compound 7(20.0mg) was obtained as a white solid.
A mixture of compound 7(20.0mg, 1.0eq) and compound 75(13.6mg, 1.0eq) was dissolved in DMF (1.0mL) and the mixture was stirred at 15 ℃ for 8 h. When the click reaction was complete, the mixture was purified by preparative HPLC. Fractions with the desired m/z (e.g., 1644.9, 1234.2, 987.6, 823.2, etc.) were lyophilized to give I-24 as a white solid (18.1mg, 53.9% yield, 98.3% purity). And (3) purification conditions:
Figure BDA0003526174380003221
Exemplary preparation of I-25, I-26, I-28 and I-29.
Figure BDA0003526174380003231
Figure BDA0003526174380003241
Figure BDA0003526174380003251
Figure BDA0003526174380003261
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.3mmol, 0.30g, 1.0mmol/g) and Fmoc-Thr (tBu) -OH (0.119g, 0.30mmol, 1.00eq) in DCM (5mL) and at 15 ℃ under N2Mix for 2 hours with bubbling. MeOH (0.3mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Trt) -OH (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. Monitoring the coupling reaction by ninhydrin tests, e.g.If it appears colorless, then the coupling is complete. If it shows a blue or reddish-brown color, the coupling is repeated and then checked again with the ninhydrin test until completion. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (10mL) × 5, MeOH (10mL) × 5 and then dried in vacuo.
Figure BDA0003526174380003271
Figure BDA0003526174380003281
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (92.5% TFA/2.5% TIS/2.5% H) at room temperature2O/2.5% 3-mercaptopropionic acid) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm).
4) The precipitate was washed two more times with cold isopropyl ether and the crude peptide was dried under vacuum for 2 hours.
5) To the crude material in ACN/H2To the mixture in O (300mL) NaHCO was added3To adjust the pH to 8, and then the mixture was stirred at 15 ℃ for 30 minutes. The mixture was quenched with 1M HCl to adjust the pH to 7. The mixture was dried via lyophilization and the residue was directly purified by preparative HPLC (TFA conditions) to give compound 2(50mg, 90% purity).
To compound 2(50mg, 10.5. mu. mmol, 1.0eq) in MeCN/H2To a mixture in O (1/1, 3mL) was added TCEP (12.0mg, 42.0. mu. mol, 4.0eq) with saturated NaHCO3The aqueous solution was basified to pH 8 and the mixture was stirred at 15 ℃ for 3 hours. The mixture was directly purified by Flash (acidic conditions, TFA) to give compound 3(30mg) as a white solid.
Compound 3(25mg, 5.48. mu. mol, 1.0eq) was dissolved in ACN (10mL) and H at 20 deg.C2O (10 mL). And then by NaHCO3The pH was adjusted to 8. The mixture was stirred at 20 ℃ for 72 hours. LCMS showed reaction completion. The solution was directly purified by preparative HPLC (TFA conditions) to give I-28 as a white solid (5.0mg, 18.97% yield, 94.8% purity). And (3) purification conditions:
Figure BDA0003526174380003282
i-25, I-26 and I-29 were prepared in a similar manner. Some results from some preparations are provided below:
compound (I) LC/MS (+ ESI; main peak) Purity (HPLC)
I-25 1491.2 95%
I-26 1461.7 96%
I-28 1520.5 95%
I-29 1432.4 95%
Exemplary preparation of I-27.
Figure BDA0003526174380003291
Figure BDA0003526174380003301
Figure BDA0003526174380003311
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.5mmol, 0.50g, 1.0mmol/g) and Fmoc-Thr (tBu) -OH (0.198g, 0.50mmol, 1.00eq) in DCM (5mL) and at 15 ℃ under N 2Mix for 2 hours with bubbling. MeOH (0.5mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Trt) -OH (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. By ninhydrin assayAn attempt was made to monitor the coupling reaction and if it appeared colorless, the coupling was complete. If it shows a blue or reddish-brown color, the coupling is repeated and then checked again with the ninhydrin test until completion. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (10mL) × 5, MeOH (10mL) × 5 and then dried in vacuo.
Figure BDA0003526174380003321
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (95% TFA/2.5% TIS/2.5% H) at room temperature2O, 10mL) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (50mL) and centrifuged (3 min at 3000 rpm).
4) Two more washes with isopropyl ether were performed, and the crude peptide was dried under vacuum for 2 hours.
5) Compound 1(800mg, crude material) was obtained as a white solid.
To compound 1 in MeCN/H2To the mixture in O (500mL) was added 0.1M I dropwise2AcOH until a pale yellow color remained, and the mixture was stirred at 15 ℃ for 5 minutes. With 0.1M Na2S2O3The mixture was quenched dropwise until the pale yellow color disappeared. The mixture was dried via lyophilization. The residue was directly purified by preparative HPLC (TFA conditions) to give compound 2(200mg, 90.0% purity, 18.2% yield).
Another exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: to a mixture containing CTC-containing resin (1.0mmol, 1.0g, 1.0mmol/g) and Fmoc-PEG10-CH2CH2DIEA (4.00eq) was added dropwise to a container of COOH (0.75g, 1.0mmol, 1.00eq) in DCM (10mL) and N at 15 deg.C 2Mix for 2 hours with bubbling. MeOH (1.0mL) was then added and the reaction solution was quenched with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (20mL) × 5. DMF (20mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (20mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Mmt) -OH (2.00eq), HBTU (1.90eq) in DMF (5mL) was added to the resin with bubbling. DIEA (4.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it shows a blue or reddish-brown color, the coupling is repeated and then checked again with the ninhydrin test until completion. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) Coupling of the last position: a solution of 2-bromoacetic acid (4.00eq) and DIC (4.00eq) was added to the resin and the mixture was taken up with N2Bubbling for 20 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) by 5, MeOH (10mL) by 5 and then dried in vacuo.
Figure BDA0003526174380003331
Figure BDA0003526174380003341
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (2% TFA/2% TIS/96% DCM, 100mL) at room temperature with N2Bubbling for 20 minutes.
2) Filtered and the filtrate collected. The clear solution was lysis buffer (100mL) containing compound 3(1.0mmol) and was used directly in the next step.
Compound 3(1.0mmol, 100mL in lysis buffer) was diluted with MeOH (1000mL), basified with TEA under an atmosphere of N2 to pH 8, then stirred at 15 ℃ for 2 hours. The solvent was removed under reduced pressure and the residue wet-milled in 0.1M HCl (cold, 100 mL). After filtration, the solid is treated with H2O (50mL) was washed and then stirred in isopropyl ether (50mL) for 10 min. Finally, the solid was dried under reduced pressure to give compound 4(1.80g, crude material).
A mixture of compound 4(1.80g, 0.63mmol, 1.00eq), compound 4a (522mg, 3.15mmol, 5.00eq), EDCI (361mg, 1.89mmol, 3.00eq) in DMF (10mL) was stirred at 15 ℃ for 3 h. The mixture was added to 0.5M HCl (cold, 100mL) and a large amount of white solid appeared. After filtration, the solid is treated with H 2O (20mL) was washed and dried via lyophilization to give compound 5(2.0g, crude material) as a white solid.
A mixture of compound 5(2.0g, crude material) was dissolved in a solution containing TFA (45.6g, 400mmol, 30mL), H2O (0.75g, 41.6mmol, 0.75mL) and triisopropylsilane (0.58g, 3.67mmol, 0.75mL), and stirred at 15 ℃ for 1 hour. The mixture was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm). The precipitate was washed two more times with cold isopropyl ether (50 mL). The crude peptide was dried in vacuo for 2 hours and then passed through flash C18: (
Figure BDA0003526174380003342
120g
Figure BDA0003526174380003343
C18 flash column, eluent: 0 to 90% MeCN/H2O gradient, 75mL/min) gave compound 6(180mg, 77.4 μmol, 11.5% yield, 90.0% purity) as a white solid.
A mixture of compound 6(80.0mg, 34.4. mu. mol, 1.00eq), compound 2(75.0mg, 34.4. mu. mol, 1.00eq), DIEA (35.5mg, 275. mu. mol, 47.9. mu.L, 8.00eq) in DMF (0.5mL) was stirred at 15 ℃ for 1 hour. The solution was directly purified by preparative HPLC (TFA conditions) to give compound I-27(67.0mg, 15.3 μmol, 44.4% yield, 95.3% purity) as a white solid. LCMS: 1451.6(+ ESI scan; main peak).
And (3) purification conditions:
Figure BDA0003526174380003351
exemplary preparation of I-30 and I-31.
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: into a vessel containing Rink amide MBHA resin (0.2mmol, 0.65g, 0.31mmol/g) in DMF (5mL) at 15 deg.C with N2Bubbling for 30 minutes. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Thr (tBu) -OH (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it appears blue or reddish-brown, the coupling is repeated and checked again with ninhydrin test until complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (10mL) × 5, MeOH (10mL) × 5 and then dried in vacuo.
After cycle 27, the Fmoc group remained on the resin, and then the Mmt group on the Cys side chain was removed by 2% TFA/2% TIS/DCM (2min × 5), and then a disulfide bond was formed on the resin by adding 2eq NCS and allowing to react for 15 min. Disulfide formation was confirmed by pilot cleavage and LCMS analysis, and then Fmoc was removed and 2-bromoacetic acid was coupled as the last residue.
Figure BDA0003526174380003361
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (92.5% TFA/2.5% TIS/2.5% H) at room temperature2O/2.5% 3-mercaptopropionic acid, 10mL) and stirred for 2 hours.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm).
4) The precipitate was washed twice more with isopropyl ether and the crude peptide was dried under vacuum for 2 hours to give the crude peptide as a white solid (0.68g, crude material).
5) Crude peptide (0.68g) was dissolved in MeCN/H2O (200mL), then 0.2M NaHCO 3The pH was adjusted to 8 and stirred at 15 ℃ for 1 hour. After the reaction was complete, the pH of the mixture was adjusted to 6 by 1M HCl. The mixture was dried by lyophilization. By preparative HPLC (acid)Sexual conditions, TFA) to afford compound 425(6mg, 0.89% yield, 96.6% purity) as a white solid.
And (3) purification conditions:
Figure BDA0003526174380003371
i-31 was prepared in a similar manner. Some results from some preparations are provided below:
compound (I) LC/MS (+ ESI; main peak) Purity (HPLC)
I-30 1088.3 91%
I-31 1113.4 95%
Exemplary preparation of I-32.
The preparation of I-32 is described below as an example.
Figure BDA0003526174380003372
Figure BDA0003526174380003381
Figure BDA0003526174380003391
Figure BDA0003526174380003401
A mixture of compound 1a (1g, 5.49mmol, 1eq), compound 1(1.50g, 3.53mmol, 0.6eq), TEA (1.11g, 10.98mmol, 1.53mL, 2eq) in EtOH (20mL) was stirred at 90 ℃ for 16 h. The solvent was removed under reduced pressure. The residue was diluted with DCM (100mL), washed with 1M HCl (20mL), and dried over anhydrous Na2SO4Drying and concentration under reduced pressure gave compound 2 as a brown oil (2.5g, 4.25mmol, 77.3% yield).
Compound 2(2.5g, 4.25mmol, 1eq) in TFA (10mL) and DCM (10mL) was stirred at 15 ℃ for 0.5 h. The solvent was removed under reduced pressure. By FLASH C18(
Figure BDA0003526174380003411
120g
Figure BDA0003526174380003412
C18 flash column, eluent: 0 to 90% MeCN/H2O ether gradient, 75mL/min) to afford compound 3 as a brown oil (1.8g, 2.99mmol, 70.3% yield, TFA).
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (2.0mmol, 2.0g, 1.0mmol/g) and Fmoc-Lys (Boc) -OH (936mg, 2.0mmol, 1.00eq) in DCM (20mL) and was added N at 15 deg.C2Mix for 2 hours with bubbling. MeOH (2mL) was then added and washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (40mL) × 5. DMF (40mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (20mL) 5 and then proceeded to the next stepAnd (5) carrying out a step.
2) Coupling: a mixture of tert-butyl 3- (2- (2-aminoethoxy) ethoxy) propionate (2.00eq), CDI (2.0eq) in DMF (5mL) was stirred at 15 ℃ for 1 hour. The resulting mixture and DMAP (0.4eq) were then added to the resin with N at 15 deg.C2Bubbling for 72 hours. The coupling reaction was monitored by ninhydrin test and it appeared colorless, indicating that the coupling was complete. The resin was then washed with DMF (20mL) × 5, MeOH (20mL) × 5 and then dried in vacuo.
Numbering Material Coupling reagent
1 Fmoc-Lys(Boc)-OH(1.00eq) DIEA(4.00eq)
2 3- (2- (2-Aminoethoxy) ethoxy) propionic acid tert-butyl ester (2.00eq) CDI(2.00eq),DMAP(0.4eq)
Peptide cleavage and purification:
1) To the flask containing the side chain protected peptide was added lysis buffer (20% HFIP/DCM, 40mL) at room temperature and stirred twice for 30 min.
2) Filtered and the filtrate collected.
3) The combined filtrates were concentrated under reduced pressure.
4) By Flash C18 (neutral condition, H)2O/MeCN) to give a colorless oilCompound 5(500mg, crude material).
A mixture of compound 3(519mg, 1.06mmol, 1.07eq), compound 5(0.5g, 988. mu. mol, 1.0eq), DIEA (383mg, 2.97mmol, 516.76. mu.L, 3.0eq), HBTU (375mg, 988. mu. mol, 1.0eq) in DMF (20mL) was stirred at 15 ℃ for 1 hour. By FLASH C18(
Figure BDA0003526174380003413
120g
Figure BDA0003526174380003414
C18 flash column, eluent: 0 to 90% MeCN/H2O ether gradient, 75mL/min) to afford compound 6 as a colorless oil (0.6g, 614.63 μmol, 62.15% yield).
Compound 6(0.6g, 614 μmol, 1eq) was dissolved in TFA (6mL) and DCM (6mL) and stirred at 15 ℃ for 0.5 h. The solvent was removed under reduced pressure. By FLASH C18(
Figure BDA0003526174380003421
120g
Figure BDA0003526174380003422
C18 flash column, eluent: 0 to 90% MeCN/H2O ether gradient, 75mL/min) to give compound 7 as a colorless oil (500mg, 535 μmol, 87.1% yield, TFA salt).
A mixture of compound 7(500mg, 535. mu. mol, 1.0eq, TFA), Fmoc-OSu (189mg, 562. mu. mol, 1.05eq), DIEA (207mg, 1.61mmol, 279. mu.L, 3.0eq) in DMF (5mL) was stirred at 15 ℃ for 1 h. By FLASH C18(
Figure BDA0003526174380003423
120g
Figure BDA0003526174380003424
C18 flash column, eluent: 0 to 90% MeCN/H2O ether gradient, 75mL/min) to give compound 8 as a white solid (350mg, 335 μmol, 62.7% yield).
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.4mmol, 0.4g, 1.0mmol/g, 1.2eq) and Compound 8(350mg, 335. mu. mol, 1.0eq) in DCM (3mL) and was treated at 15 ℃ under N2Mix for 2 hours with bubbling. MeOH (0.3mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Glu-OtBu (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it appears blue or reddish-brown, the coupling is repeated and checked again with ninhydrin test until complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection of Fmoc: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating step 2 for the next amino acid: (No. 3, 16- (tert-butoxy) -16-oxohexadecanoic acid in Table 2).
5) Deprotection of Dde: will contain 3% of N2H4.H2DMF of O (10mL) was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction was monitored by ninhydrin test and it showed blue or reddish-brown indicating that the reaction was complete.
6) Repeating steps 2 and 3 for all other amino acids: see below.
7) Last one isCoupling of individual positions: a solution of 2-bromoacetic acid (4.00eq) and DIC (4.00eq) was added to the resin and the mixture was taken up with N2Bubbling for 20 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) by 5, MeOH (10mL) by 5 and then dried in vacuo.
Numbering Material Coupling reagent
1 Compound 8(1.0eq) DIEA(4.0eq)
2 Fmoc-Glu-OtBu(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
3 16- (tert-butoxy) -16-oxohexadecanoic acid (2.0eq) HATU (1.90eq) and DIEA (4.0eq)
4 Fmoc-Cys(Mmt)-OH(2.0eq) HATU (1.90eq) and DIEA (4.0eq)
5 Fmoc-Asp(OtBu)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
6 Fmoc-HomNle-OH(2.0eq) HATU (1.90eq) and DIEA (4.0eq)
7 Fmoc-Bip-OH(2.0eq) HATU (1.90eq) and DIEA (4.0eq)
8 Fmoc-Leu-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
9 Fmoc-Val-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
10 Fmoc-Gly-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
11 Fmoc-Asp(OtBu)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
13 Fmoc-His(Trt)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
13 Fmoc-Tyr(tBu)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
14 Fmoc-HomNle-OH(2.0eq) HATU (1.90eq) and DIEA (4.0eq)
15 Fmoc-Arg(Pbf)-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
16 Fmoc-Ala-OH(3.0eq) HBTU (2.85eq) and DIEA (6.0eq)
17 2-Bromoacetic acid (4.0eq) DIC(2.0eq)
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (2% TFA/2% Tis/96% DCM, 40mL) at room temperature with N2Bubbling for 20 minutes.
2) Filtered and the filtrate collected. The clear solution was lysis buffer (400mL) containing compound 10(335 μmol) and was used directly in the next step.
Compound 10 (335. mu. mol, 400mL in lysis buffer) was diluted with MeOH (400mL) in N2Basified with TEA under atmosphere to pH 8 and then stirred at 15 ℃ for 2 h. The solvent was removed under reduced pressure and the residue wet-milled in 0.1M HCl (cold, 100 mL). After filtration, the solid is treated with H2O (50mL) was washed and then stirred in isopropyl ether (50mL) for 10 min. Finally, the solid was dried under reduced pressure to give compound 11(1.0g, crude material).
Mixing Compound 11(650mg, 186. mu. mol, 1.0eq), 2, 3, 5, 6-tetrafluorophenol (155mg, 934. mu. mol, 5.0eq), EDCI (107mg, 560. mu. mol, 3.0eq) in DMF (6mL)The mixture was stirred at 15 ℃ for 3 hours. The mixture was added to 0.5M HCl (cold, 50mL), filtered, and the solid was washed with H2O (cold, 50mL), isopropyl ether (50mL) were washed, dried via lyophilization to give compound 12(700mg, crude material) as a white solid.
With TFA (15.40g, 135mmol, 10mL), H2A deprotected mixture of O (250mg, 13.88mmol, 0.25mL) and triisopropylsilane (192.75mg, 1.22mmol, 0.25mL) treated Compound 12(600mg, 165. mu. mol) and stirred at 15 ℃ for 1 h. The mixture was precipitated with cold isopropyl ether (50mL) and centrifuged (3 min at 3000 rpm). Two more washes (50mL) were performed with isopropyl ether. The crude peptide was dried in vacuo for 2 hours. The solution was directly purified by preparative HPLC (TFA conditions) to give compound 13(25mg, 8.60 μmol, 5.2% yield) as a white solid.
Figure BDA0003526174380003441
Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: into a vessel containing Rink amide MBHA resin (0.2mmol, 0.65g, 0.31mmol/g) in DMF (5mL) at 15 deg.C with N 2Bubbling for 30 minutes. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Thr (tBu) -OH (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. Monitoring by ninhydrin testThe deprotection reaction is detected and if it shows a blue or red-brown colour, the reaction is complete.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (10mL) × 5, MeOH (10mL) × 5 and then dried in vacuo.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(2.00eq) HBTU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
14 Fmoc-Asp(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
Peptide cleavage, cyclization and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (95% TFA/2.5% TIS/2.5% H) at room temperature2O, 10mL) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm).
4) Washed twice more with isopropyl ether and the crude peptide was dried under vacuum for 2 hours to give compound 14(0.2mmol, crude material).
5) Dissolving Compound 14 in MeCN/H2O (1: 1, 200mL), and then 0.1M I was added dropwise2HOAc until it remains pale yellow. After 10 minutes, use 0.1M Na2S2O3The mixture was quenched dropwise until the pale yellow color disappeared. The mixture was dried via lyophilization and the residue was directly purified by preparative HPLC (TFA conditions) to give compound 15(120mg, 90% purity).
Figure BDA0003526174380003461
A mixture of compound 11(25mg, 8.7. mu. mol, 1.0eq), compound 15(17.4mg, 10.5. mu. mol, 1.2eq), DIEA (11.3mg, 87.7. mu. mol, 15.3. mu.L, 10eq) in DMF (0.2mL) was stirred at 15 ℃ for 1 hour. The solution was directly purified by preparative HPLC (TFA conditions) to give I-32(8.9mg, 1.89 μmol, 21.5% yield, 92% purity) as a white solid. LCMS: 1443.4(+ ESI scan; main peak). And (3) purification conditions:
Figure BDA0003526174380003462
exemplary preparation of I-38.
Figure BDA0003526174380003471
Figure BDA0003526174380003481
Figure BDA0003526174380003491
The chemical formula is as follows: c210H295N49O48S3
Figure BDA0003526174380003501
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: into a vessel containing Rink amide MBHA resin (0.2mmol, 0.65g, 0.31mmol/g) in DMF (5mL) at 15 deg.C with N2Bubbling for 30 minutes. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Thr (tBu) -OH (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. Tree (R)The lipids were then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (10mL) × 5, MeOH (10mL) × 5 and then dried in vacuo.
Note that: 3% N2H4.H2O/DMF was used for Dde deprotection.
Figure BDA0003526174380003511
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (95% TFA/2.5% TIS/2.5% H) at room temperature2O, 10mL) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm).
4) Two more washes with isopropyl ether were performed, and the crude peptide was dried under vacuum for 2 hours.
5) To compound 1 in MeCN/H2To the mixture in O (200mL) was added 0.1M I dropwise2HOAc until it remains pale yellow, followed by 0.1M Na2S2O3The mixture was quenched dropwise until the pale yellow color disappeared. The mixture was dried via lyophilization. The residue was purified directly by preparative HPLC (TFA conditions) to give compound 2(125mg) as a white solid.
Another exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.5mmol, 0.5g, 1.0mmol/g) and 5- ((((9H-fluoren-9-yl) methoxy) carbonyl) amino) pentanoic acid (0.17g, 0.5mmol, 1.00eq) in DCM (5mL) and at 15 ℃ in N2Mix for 2 hours with bubbling. MeOH (0.5mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (20mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Mmt) -OH (2.00eq), HBTU (1.90eq) in DMF (5mL) was added to the resin with bubbling. DIEA (4.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) Coupling of the last position: a solution of 2-bromoacetic acid (4.00eq) and DIC (4.00eq) was added to the resin and the mixture was taken up with N2Bubbling for 20 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) by 5, MeOH (10mL) by 5 and then dried in vacuo.
Figure BDA0003526174380003521
Figure BDA0003526174380003531
Peptide cleavage and purification:
3) to a flask containing the side chain protected peptide was added lysis buffer (2% TFA/2% Tis/96% DCM, 50mL) at room temperature with N2Bubbling for 20 minutes.
4) Filtered and the filtrate collected. The clear solution was lysis buffer (50mL) containing compound 3(0.5mmol) and was used directly in the next step.
Compound 3(0.5mmol, 50mL in lysis buffer) was diluted with MeOH (500mL) in N2Basified with TEA under atmosphere to pH 8 and then stirred at 15 ℃ for 2 h. The solvent was removed under reduced pressure. The residue was wet-milled in 0.1M HCl (cold, 50mL), filtered, and the solid was washed with H2O (50mL) was washed and then stirred in isopropyl ether (50mL) for 10 min. Finally, the solid was dried under reduced pressure to give compound 4(1.00g, crude material).
A mixture of compound 4(1.00g, 0.41mmol, 1.00eq), compound 4a (340mg, 2.06mmol, 5.00eq), EDCI (236mg, 1.23mmol, 3.00eq) in DMF (6mL) was stirred at 15 ℃ for 3 h. The mixture was added to 0.5M HCl (cold, 60mL) and a large amount of white solid appeared. After filtration, the solid is treated with H 2O (50mL) was washed and dried via lyophilization to give compound 5(1.1g, crude material) as a white solid.
With TFA (22.8g, 200mmol, 15mL), H2A solvent mixture of O (0.38g, 20.8mmol, 0.38mL) and triisopropylsilane (0.29g, 1.83mmol, 0.38mL) treated Compound 5(1.1g, crude) and was stirred at 15 deg.C for 1 hour. The mixture was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm). The precipitate was washed two more times with cold isopropyl ether (50 mL). The crude peptide was dried in vacuo for 2 hours. By FLASH C18(
Figure BDA0003526174380003532
120g
Figure BDA0003526174380003533
C18 flash column, eluent: 0 to 90% MeCN/H2O gradient, 75mL/min) gave compound 6(90mg, 47.0 μmol, 11.0% yield, 90.0% purity) as a white solid.
A mixture of Compound 6(40.0mg, 20.9. mu. mol, 1.00eq), Compound 2(43.8mg, 16.73. mu. mol, 0.80eq), DIEA (21.6mg, 167. mu. mol, 29.1. mu.L, 8.00eq) in DMF (0.5mL) was stirred at 15 ℃ for 1 hour. The solution was directly purified by preparative HPLC (acidic conditions, TFA) to give Compound I-38 (Q1: 46.3mg, 10.2. mu. mol, 96.3% purity) and Compound I-38 (Q2: 20.9mg, 4.6. mu. mol, 85.4% purity) as white solids. LCMS: 874.9(+ ESI scans; main peaks; also 1457.6, 1093.3, 729.5, etc.).
And (3) purification conditions:
Figure BDA0003526174380003541
exemplary preparation of I-39.
Figure BDA0003526174380003542
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.5mmol, 0.50g, 1.0mmol/g) and Fmoc-Thr (tBu) -OH (0.198g, 0.50mmol, 1.00eq) in DCM (5mL) and at 15 ℃ under N2Mix for 2 hours with bubbling. MeOH (0.5mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Trt) -OH (3.00eq), HBTU (2.85eq) in DMF (5mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it appears blue or reddish-brown, the coupling is repeated and checked again with ninhydrin test until complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was mixed at 15 deg.C N for things2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) Urea at the last position: a mixture of Boc-NH2-NH2(6.00eq), CDI (6.00eq) and TEA (6.00eq) in DMF (5mL) was stirred at 15 ℃ for 1 hour. The mixture and DMAP (6.00eq) were then added to the resin with N2Bubbling for 48 hours. The reaction was monitored by ninhydrin test and if it showed no colour, the coupling was complete. The resin was then washed with DMF (10mL) by 5, MeOH (10mL) by 5 and then dried in vacuo.
Figure BDA0003526174380003551
Peptide cleavage, cyclization and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (95% TFA/2.5% TIS/2.5% H) at room temperature2O, 10mL) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm).
4) Two more washes with isopropyl ether were performed, and the crude peptide was dried under vacuum for 2 hours.
5) Dissolving Compound 1 in MeCN/H2O (500mL), and then 0.1M I was added dropwise 2HOAc until it remains pale yellow. After 10 minutes, use 0.1M Na2S2O3The mixture was quenched dropwise until the pale yellow color disappeared. The mixture was then dried via lyophilization. Finally, the residue was directly purified by preparative HPLC (acidic conditions, TFA) to give compound 2(65mg, 90% purity).
Figure BDA0003526174380003561
Another exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: to a mixture containing CTC-containing resin (1.0mmol, 1.0g, 1.0mmol/g) and Fmoc-PEG10-CH2CH2DIEA (4.00eq) was added dropwise to a container of COOH (0.75g, 1.0mmol, 1.00eq) in DCM (10mL) and N at 15 deg.C2Mix for 2 hours with bubbling. MeOH (1.0mL) was then added and the reaction solution was quenched with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (20mL) × 5. DMF (20mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (20mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Mmt) -OH (2.00eq), HBTU (1.90eq) in DMF (5mL) was added to the resin with bubbling. DIEA (4.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it appears blue or reddish-brown, the coupling is repeated and checked again with ninhydrin test until complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: (2 to 14 in Table 2).
5) Coupling of the last position: a solution of 2-bromoacetic acid (4.00eq) and DIC (4.00eq) was added to the resin and the mixture was taken up with N2Bubbling for 20 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) by 5, MeOH (10mL) by 5 and then dried in vacuo.
Figure BDA0003526174380003571
Figure BDA0003526174380003581
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (2% TFA/2% Tis/96% DCM, 100mL) at room temperature with N2Bubbling for 20 minutes.
2) Filtered and the filtrate collected. The clear solution was lysis buffer (100mL) containing compound 4(1.0mmol) and was used directly in the next step.
Figure BDA0003526174380003582
Compound 4(1.0mmol, 100mL in lysis buffer) was diluted with MeOH (1000mL), basified with TEA under an atmosphere of N2 to pH 8, then stirred at 15 ℃ for 2 hours. The solvent was removed under reduced pressure and the residue wet-milled in 0.1M HCl (cold, 100 mL). After filtration, the solid is treated with H 2O (50mL) was washed and then stirred in isopropyl ether (50mL) for 10 min. Finally, the solid was dried under reduced pressure to give compound 5(1.80g, crude material).
Figure BDA0003526174380003591
A mixture of compound 5(1.80g, 0.63mmol, 1.00eq), compound 4a (522mg, 3.15mmol, 5.00eq), EDCI (361mg, 1.89mmol, 3.00eq) in DMF (10mL) was stirred at 15 ℃ for 3 h. The mixture was added to 0.5M HCl (cold, 100mL) and a large amount of white solid appeared. After filtration, the solid is treated with H2O (20mL) was washed and dried via lyophilization to give compound 6(2.0g, crude material) as a white solid.
Figure BDA0003526174380003592
Compound 6(2g, crude material) in TFA (45.6g, 400mmol, 30mL), H2A mixture of O (0.75g, 41.6mmol, 0.75mL) and triisopropylsilane (0.58g, 3.67mmol, 0.75mL) was stirred at 15 ℃ for 1 hour. The mixture was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm). Two more washes (50mL) were performed with isopropyl ether. The crude peptide was dried in vacuo for 2 hours. By fast C18(
Figure BDA0003526174380003601
120g
Figure BDA0003526174380003602
C18 flash column, eluent: 0 to 90% MeCN/H2O gradient, 75mL/min) gave compound 7(180mg, 77.4 μmol, 11.5% yield, 90.0% purity) as a white solid.
Figure BDA0003526174380003603
A mixture of compound 7(47mg, 20.22. mu. mol, 1.00eq), compound 2(40mg, 20.82. mu. mol, 1.0eq), DIEA (20.9mg, 161.7. mu. mol, 28.1. mu.L, 8eq) in DMF (0.2mL) was stirred at 15 ℃ for 30 minutes. The solution was directly purified by preparative HPLC (TFA conditions) to give I-39 (Q1: 3.7mg, 0.83. mu. mol, 4.0% yield, 94% purity) and I-39 (Q2: 9.8mg, 2.19. mu. mol, 10.6% yield, 92% purity) as white solids. LCMS: 1362.2(+ ESI scan; main peak).
And (3) purification conditions:
Figure BDA0003526174380003604
Figure BDA0003526174380003611
exemplary preparation of I-40.
Figure BDA0003526174380003612
Figure BDA0003526174380003621
Figure BDA0003526174380003631
Figure BDA0003526174380003641
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: a vessel containing Rink amide MBHA resin (0.5mmol, 1.61g, 0.31mmol/g) in DMF (10mL) was bubbled with N2 for 30 minutes at 15 ℃. DMF (20mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (20mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Trt) -OH (3.00eq), HBTU (2.85eq) in DMF (10mL) was added to the resin with bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C 2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (20mL) × 5.
3) Deprotection: DMF (20mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (20mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (20mL) × 5, MeOH (20mL) × 5 and then dried in vacuo.
Figure BDA0003526174380003642
Figure BDA0003526174380003651
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (95% TFA/2.5% TIS/2.5% H) at room temperature2O, 20mL) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (50mL) and centrifuged (3 min at 3000 rpm).
4) Two more washes with isopropyl ether were performed, and the crude peptide was dried under vacuum for 2 hours.
5) To the crude material in ACN/H2To the mixture in O (300mL) was added 0.1M I dropwise2AcOH until a pale yellow color remained, and the mixture was stirred at 15 ℃ for 5 minutes. With 0.1M Na 2S2O3The mixture was quenched dropwise until the pale yellow color disappeared. The mixture was dried via lyophilization, and the residue was then directly purified by preparative HPLC (acidic conditions, TFA) to give compound 2(150mg, 90% purity)
Another exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.5mmol, 0.50g, 1.0mmol/g) and Fmoc-Gly-OH (0.149g, 0.50mmol, 1.00eq) in DCM (5mL) and was added N at 15 deg.C2Mix for 2 hours with bubbling. MeOH (0.5mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5, and then DMF (10mL) with 20% piperidine was added, and the mixture was washed with N at 15 ℃2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. Resin compositionWash with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Mmt) -OH (2.00eq), HBTU (1.90eq) in DMF (5mL) was added to the resin with bubbling. DIEA (4.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it shows a blue or reddish-brown color, the coupling is repeated once more and checked again with ninhydrin test until the coupling is complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: (2 to 14 in Table 2).
5) Coupling of the last position: a solution of 2-bromoacetic acid (4.00eq) and DIC (4.00eq) was added to the resin and the mixture was taken up with N2Bubbling for 20 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (10mL) by 5, MeOH (10mL) by 5 and then dried in vacuo.
Figure BDA0003526174380003661
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (2% TFA/2% TIS/96% DCM, 100mL) at room temperature with N2Bubbling for 20 minutes.
2) Filtered and the filtrate collected. The clear solution was lysis buffer containing compound 3(0.5mmol, 100mL) and was used directly in the next step.
3) Compound 3(0.5mmol, 100mL in lysis buffer) was diluted with MeOH (500mL), basified with TEA to pH 8 under N2 atmosphere, and ligated Followed by stirring at 15 ℃ for 4 hours. The solvent was removed under reduced pressure and the residue wet-milled in 0.1M HCl (cold, 100 mL). After filtration, the solid is treated with H2O (50mL) was washed, and then stirred in isopropyl ether (50mL) for 10 minutes, and finally dried under reduced pressure to give compound 4(1.2g, crude material).
A mixture of compound 4(1.2g, 1.00eq), compound 4a (501mg, 6.00eq), EDCI (288mg, 3.00eq) in DMF (10mL) was stirred at 15 ℃ for 3 hours. The mixture was then added to 0.5M HCl (cold, 100mL) and a large amount of white solid appeared. After filtration, the solid is treated with H2O (20mL) was washed and dried via lyophilization to give compound 5(1.8g, crude material) as a white solid. Deprotection mixture (95% TFA/2.5% TIS/2.5% H)2O, 30mL) was treated with the crude material for 1.5 hours. The mixture was precipitated with cold isopropyl ether (100mL) and centrifuged (3 min at 3000 rpm). The precipitate was washed two more times with cold isopropyl ether (50 mL). The crude peptide was dried in vacuo for 2 hours. By fast C18(
Figure BDA0003526174380003673
120g
Figure BDA0003526174380003674
C18 flash column, eluent: 0 to 90% MeCN/H2O gradient, 75mL/min) the residue was purified directly to give compound 6(55mg) as a white solid.
A mixture of compound 6(55mg, 1.00eq), compound 2(51.3mg, 1.10eq), DIEA (30.7. mu.L, 6.00eq) in DMF (0.5mL) was stirred at 15 ℃ for 1 hour. The solution was directly purified by preparative HPLC (TFA conditions) to give compound 523(15.4mg, 15.9% yield, 98.6% purity) as a white solid. LCMS: 1098.4(+ ESI scan; main peak).
And (3) purification conditions:
Figure BDA0003526174380003671
exemplary preparation of I-33, I-34, I-35, I-36, I-37, I-41 and I-42.
Figure BDA0003526174380003672
Exemplary peptide synthesis. Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: DIEA (4.00eq) was added dropwise to a vessel containing CTC resin (0.5mmol, 0.50g, 1.0mmol/g, 1.00eq) and Fmoc-Thr (tBu) -OH (0.158g, 0.40mmol, 0.8eq) in DCM (5mL) and at 15 ℃ under N2Mix for 2 hours with bubbling. MeOH (0.5mL) was then added and the reaction solution was washed with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: a solution of Fmoc-Cys (Trt) -OH (877mg, 1.5mmol, 3.00eq), HBTU (541mg, 1.42mmol, 2.85eq) in DMF (5mL) was added to the resin with N2 bubbling. DIEA (6.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it appears blue or reddish-brown, the coupling is repeated and checked again with ninhydrin test until complete. The resin was then washed with DMF (10mL) × 5.
3) Deprotection: DMF (10mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (10mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) After completion of the last position, the resin was washed with DMF (10mL) × 5, MeOH (10mL) × 5 and then dried in vacuo.
Numbering Material Coupling reagent
1 Fmoc-Thr(tBu)-OH(0.80eq) DIEA(4.00eq)
2 Fmoc-Cys(Trt)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
3 Fmoc-Trp(Boc)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
4 Fmoc-Val-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
5 Fmoc-Leu-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
6 Fmoc-Glu(OtBu)-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
7 Fmoc-Gly-OH(3.00eq) HBTU (2.85eq) and DIEA (6.00eq)
8 Fmoc-Leu-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
9 Fmoc-His(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
10 Fmoc-Trp(Boc)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
11 Fmoc-Ala-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
12 Fmoc-Cys(Trt)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
13 Fmoc-Asp(OtBu)-OH(3.00eq) HATU (2.85eq) and DIEA (6.00eq)
14 Fmoc-PEG8-CH2CH2COOH(2.00eq) HATU (1.90eq) and DIEA (4.00eq)
Peptide cleavage and purification:
1) to the containing side at room temperatureChain protected peptide flasks lysis buffer (95% TFA/2.5% TIS/2.5% H) was added2O) and stirred for 1 hour.
2) Filtered and the filtrate collected.
3) The peptide was precipitated with cold isopropyl ether (50mL) and centrifuged (3 min at 3000 rpm).
4) Two more washes with isopropyl ether were performed, and the crude peptide was dried under vacuum for 2 hours.
5) Compound 5(800mg, crude material) was obtained as a white solid.
Figure BDA0003526174380003691
Dissolving Compound 1 in MeCN/H2O (500mL), and 0.1M I was added dropwise2HOAc until the color of the mixture became light yellow, followed by stirring the mixture at 15 ℃ for 10 minutes. With 0.1M Na2S2O3The mixture was quenched dropwise until the color of the mixture became colorless and dried via lyophilization. The residue was directly purified by preparative HPLC (acidic conditions, TFA) to give compound 2(170mg, 90.0% purity, 17.4% yield).
Figure BDA0003526174380003692
Peptides were synthesized using standard Fmoc chemistry, for example:
1) preparing resin: to a mixture containing CTC-containing resin (0.62mmol, 0.62g, 1.0mmol/g, 1.25eq) and Fmoc-PEG8-CH2CH2DIEA (4.00eq) was added dropwise to a container of COOH (0.198g, 0.50mmol, 1.00eq) in DCM (10mL) and N at 15 deg.C2Mix for 2 hours with bubbling. MeOH (1.0mL) was then added and the reaction solution was quenched with N2Bubbling was continued for another 30 minutes. The resin was washed with DMF (10mL) × 5. DMF (10mL) containing 20% piperidine was then added and the mixture was stirred with N at 15 deg.C2Bubbling for 30 minutes. The mixture was then filtered to obtain a resin. The resin was washed with DMF (10mL) × 5 before proceeding to the next step.
2) Coupling: in N2A solution of Fmoc-Cys (Mmt) -OH (615mg, 1.00mmol, 2.00eq), HBTU (360mg, 0.95mmol, 1.90eq) in DMF (10mL) was added to the resin with bubbling. DIEA (4.00eq) was then added dropwise to the mixture and N was used at 15 deg.C2Bubbling for 30 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. If it appears blue or reddish-brown, the coupling is repeated and checked again with ninhydrin test until complete. The resin was then washed with DMF (20mL) × 5.
3) Deprotection: DMF (20mL) containing 20% piperidine was added to the resin and the mixture was taken up with N at 15 deg.C2Bubbling for 30 minutes. The resin was then washed with DMF (20mL) × 5. The deprotection reaction is monitored by the ninhydrin test and is complete if it shows a blue or brownish red colour.
4) Repeating steps 2 and 3 for all other amino acids: see below.
5) Coupling of the last position: a solution of 2-bromoacetic acid (4.00eq) and DIC (4.00eq) was added to the resin and the mixture was taken up with N2Bubbling for 20 minutes. The coupling reaction was monitored by ninhydrin test and if it appeared colorless, the coupling was complete. The resin was then washed with DMF (20mL) × 5, MeOH (20mL) × 5 and then dried in vacuo.
Figure BDA0003526174380003701
Figure BDA0003526174380003711
Peptide cleavage and purification:
1) to a flask containing the side chain protected peptide was added lysis buffer (2% TFA/2% TIS/96% DCM, 100mL) at room temperature with N2Bubbling for 20 minutes.
2) Filtered and the filtrate collected. The clear solution was lysis buffer containing compound 3(1.0mmol, 100mL) and was used directly in the next step.
Figure BDA0003526174380003712
Compound 3(1.0mmol, 100mL in lysis buffer) was diluted with MeOH (1000mL), basified with TEA under an atmosphere of N2 to pH 8, and then stirred at 15 ℃ for 2 hours. The solvent was removed under reduced pressure and the residue wet-milled in 0.1M HCl (cold, 100 mL). After filtration, the solid is treated with H2O (50mL) was washed and then stirred in isopropyl ether (50mL) for 10 min. Finally, the solid was dried under reduced pressure to give compound 4(0.75g, crude material).
Figure BDA0003526174380003713
A mixture of compound 4a (226.2mg, 1.36mmol, 5.00eq), compound 4(0.75g, 272.4. mu. mol, 1.00eq), EDCI (156.71mg, 817.48. mu. mol, 3.00eq) in DMF (5mL) was stirred at 15 ℃ for 3 hours. The mixture was added to 0.5M HCl (cold, 50mL) and a large amount of white solid appeared. After filtration, the solid is treated with H2O (20mL) was washed and dried via lyophilization to give compound 5(0.80g, crude material) as a white solid.
Figure BDA0003526174380003721
Compound 5(0.80g, crude material) was dissolved in a solution containing TFA (17.6g, 154.3mmol, 11.4mL), H2O (285.7mg, 15.8mmol, 285. mu.L) and triisopropylsilane (220.29mg, 1.39mmol, 285.71. mu.L), and the mixture was stirred at 15 ℃ for 1 hour. The mixture was then precipitated with cold isopropyl ether (100mL) and centrifuged (3 minutes at 3000 rpm). The precipitate was washed two more times with cold isopropyl ether (50 mL). The crude peptide was dried in vacuo for 2 hours. By fast C18(
Figure BDA0003526174380003722
120g
Figure BDA0003526174380003723
C18 flash column, eluent: 0 to 90% MeCN/H2O gradient, 75mL/min) gave compound 6(90mg, 40.2 μmol, 14.5% yield, 90.0% purity) as a white solid.
Figure BDA0003526174380003731
A mixture of compound 6(50mg, 22.3. mu. mol, 1.00eq), compound 2(43.60mg, 22.3. mu. mol, 1.00eq), DIEA (23.1mg, 178.7. mu. mol, 31.1. mu.L, 8.00eq) in DMF (0.5mL) was stirred at 15 ℃ for 1 hour. The solution was directly purified by preparative HPLC (acidic conditions, TFA) to give I-33(33.0mg, 7.71 μmol, 34.4% yield, 93.9% purity) as a white solid.
And (3) purification conditions:
Figure BDA0003526174380003732
i-34, I-35, I-36, I-37, I-41 and I-42 were prepared in a similar manner. Some results from some preparations are provided below:
Compound (I) LC/MS (+ ESI; main peak) Purity (HPLC)
I-33 1342.7 94%
I-34 1166.6 96%
I-35 1195.8 97%
I-36 1254.6 97%
I-37 1313.3 96%
I-41 1366.5 92%
I-42 1361.6 91%
In many cases, various other compounds, including various ARM agents, are also prepared and evaluated using one or more of the techniques described in the examples.
Example 2 the provided compounds recruit antibodies to target cells.
Various techniques can be used to assess and/or characterize the recruitment of antibodies to target cells (e.g., cancer cells). One such assay is a ternary binding assay. Exemplary schemes are described herein. Compounds were diluted in DMSO (MP 191418) into 1000 x to 96 well polypropylene plates (Corning 3357) at the starting concentration used in the assay. It was then serially diluted in 1/2 log increments to yield 8 to 12 concentrations in DMSO (depending on the assay). These DMSO stocks were then gradually diluted 1/100 into PBS (VWR cat No. 20012043). The stepwise diluted series of compounds was then added to the polypropylene assay plate at 1/10 volumes of the assay volume. Daudi (ATCC CCL-213) (non-adherent cell line) was counted and centrifuged, and resuspended in flow buffer at a concentration of 100,000 cells per 90 microliters: 1% BSA (American Bio) AB 01088-00100; 0.5mM EDTA (VWR 45001-122); PBS labeled with 11.1. mu.g/mL human IgG1K phycoerythrin (southern Biotech accession 0151K-09PE) (VWR catalog No. 20012043). Cells were then added to polypropylene plates containing stepwise diluted compounds and incubated at 37 ℃ for 30 minutes. At the end of incubation, cells were centrifuged and washed 2X with running buffer containing 0.5% Tween 20(BP 337-500). Samples were analyzed on a BD FacsCelesta. Mean fluorescence was analyzed using Graphpad Prism and a curve was fitted using log (inhibitor) and reaction-variable slope (four parameters). As shown in fig. 1, the provided compounds can recruit antibodies to target cells. In one set of experiments, IC50 for I-9 was about 16nM, and IC50 for I-17 was about 2.6 nM. The identity and/or activity of the provided technology is confirmed by assessing additional compounds. For example, various provided compounds can recruit antibodies to target cells, as shown below (MFI at various concentrations):
Figure BDA0003526174380003741
Figure BDA0003526174380003751
Figure BDA0003526174380003761
IC50(nM) from some assessments e.g.:
ID IC50(nM) ID IC50(nM)
I-9 16 I-34 20
I-17 5.9 I-35 10
I-25 9.5 I-36 10
I-26 14 I-37 7.1
I-27 6 I-38 >3000
I-28 15 I-39 3.3
I-29 61 I-40 50
I-30 14 I-42 2.2
I-31 >3000 I-41 3
I-32 88 I-43 43
I-33 9.3 I-44 19
effective recruitment of effector cells to various other cell types in the presence of IgG was confirmed by additional analysis.
As described herein, in some embodiments, a provided compound binds to CD38 with a Kd of no more than 200nM, 100nM, 50nM, 40nM, 30nM, 20nM, 10nM, or 5nM as measured by SPR and/or binds to an antibody with a Kd of no more than 200nM, 100nM, 50nM, 40nM, 30nM, 20nM, 10nM, or 5nM as measured by SPR. In some embodiments, compounds provided (e.g., I-9 and I-17) can bind to CD38 with a Kd of about 10nM and to human IgG1 and IgG2 with a Kd of about 10 to 20nM, as shown by SPR. The provided compounds are shown to bind to several IgG isotypes with nanomolar Kd binding affinity.
Example 3. the provided compounds recruit and activate effector cells.
Daudi (ATCC CCL-213) cells were resuspended in Opti-MEM (Thermo Fisher Scientific number 31985070), counted (Life Technologies Countless II cell counter) and seeded (5,000 cells per 25 microliters; corning plate: 3917). Compounds were diluted in DMSO (MP 191418) into 1000 x to 96 well polypropylene plates (corning 3357) of the starting concentration used in the assay. It was then serially diluted in 1/2 log increments to yield 8 concentrations in DMSO (depending on the assay). These DMSO stocks were then gradually diluted 1/250 into Opti-MEM. The stepwise diluted series of compounds were then added to a polypropylene assay plate at 25 μ l/well. The human antibody solution (Grifols IVIG Fleogama No. 61953-0005-2) was diluted to 40. mu.g/mL and added at 25. mu.l/well. Effector cells (ADCC reporter cells, Promega (Promega) kit: G7018) were added at 37,500 cells per 25 microliters of wells. The assay was then incubated at 37 ℃ for 18 hours. After the induction period, the plates were equilibrated to 25 ℃ followed by addition of luciferase substrate (75. mu.l/well, 1 vial in 10mL Bio-Glo assay buffer, Promega kit: G7018). Luminescence was measured (Biotek Synergy H1 microplate reader). Exemplary data from a set of experiments is presented in FIG. 2, where EC50 for I-9 is about 0.92nm and EC50 for I-17 is about 2.7 nm. Various assays, including animal models, confirm the recruitment and activation of various types of effector cells, such as macrophages, NK cells, and the like. As shown, the provided compounds can recruit and activate effector cells. Assessment of additional compounds confirms the nature and/or activity of the provided technology. For example, various provided compounds can recruit and activate effector cells (RLUs at various concentrations):
Figure BDA0003526174380003771
Figure BDA0003526174380003781
EC50(nM) from some assessments e.g.:
ID EC50(nM) ID EC50(nM)
I-9 5.5 I-34 6.7
I-17 5.9 I-35 4.4
I-25 4.7 I-36 3.4
I-26 2.4 I-37 1.8
I-27 3 I-38 53
I-28 14 I-39 0.61
I-29 9.9 I-40 50
I-30 12 I-42 1.4
I-31 >3000 I-41 24
I-32 63 I-43 11
I-33 4.3 I-44 11
example 4. the provided compounds can kill target cells.
In some embodiments, CD38-ABT ARM binds to a target cell and an antibody, e.g., a human antibody. In some embodiments, such a triplet presents an antibody to an effector cell (e.g., an NK cell) and binds to, for example, the CD16a receptor. Experiments were performed in a system similar to physiological systems using purified NK cells from multiple donors with different single nucleotide polymorphic variants of CD16a (V/V, F/F and V/F) to confirm the activity of the provided compounds (e.g., ARM). Daudi cells were transfected with KILR reporter construct (DiscoverX number 97-0002). The attenuated lentiviruses are designed to deliver Moloney Murine Leukemia Virus (MMLV) engineered to drive expression of a housekeeping gene labeled with enhanced ProLabel (ePL) (β -galactosidase (β -gal) reporter fragment). This construct is designed to remain inside the cell. Death results in the release of the KILR reporter protein into the culture medium. The detection reagent (DiscoverX number 97-0001L) contains a complementary beta-gal reporter fragment enzyme receptor (EA). The two components combine to produce a chemiluminescent signal. Data from one set of experiments is presented in FIG. 3, where EC50 for I-9 is 6.7nm and EC50 for I-17 is 5.9 nm. It has been demonstrated that provided agents, as described herein, can recruit effector cells from various sources to kill various target cells expressing CD38 associated with various conditions, disorders or diseases (e.g., cancer, e.g., multiple myeloma), which EC50 is typically in the low micromolar, nanomolar, or low nanomolar range. In some embodiments, the provided compounds can kill multiple myeloma cells in a bone marrow sample of a patient ex vivo as well as leukemia plasma cells in the blood of the patient. In some embodiments, an animal model (e.g., an animal model for burkitt's lymphoma, multiple myeloma, etc.) is utilized to confirm the efficacy of a provided compound. As shown, the provided compounds can be effective to induce killing of target cells, e.g., via recruitment of antibodies and ADCC. The identity and/or activity of the provided technology is confirmed by assessing additional compounds. For example, various provided compounds can provide activity (dead cell%) against a target cell via or including ADCC in various instances:
Figure BDA0003526174380003791
Figure BDA0003526174380003801
EC50(nM) from some assessments e.g.:
Figure BDA0003526174380003802
Figure BDA0003526174380003811
example 5 the provided compounds did not deplete effector cells expressing CD 38.
In some embodiments, the provided technology provides various advantages over existing CD 38-based technologies (e.g., technologies using CD38 antibodies). Furthermore, the provided technology does not deplete or significantly reduce normal CD 38-expressing cells, such as many immune cells, compared to CD38 antibody-based technologies. As will be appreciated by those skilled in the art, various techniques can be used to assess depletion of effector cells expressing CD 38. In one example, EasySep was used according to manufacturer's instructionsTMHuman NK cell isolation kit (STEMCELL Technologies catalog No. 17955) NK cells are purified from frozen stocks using 3X 10^7PBMC preparations (Stem cell Technologies, Inc.). NK cells were seeded in a volume of 100. mu.L per well in round bottom 96-well plates at a density of 2X 10^6 cells per ml OPTIMEM medium. Daramomum mono antibody was added to a final concentration range of 3. mu.g/mL to 0.1. mu.g/mL. Compound I-9 was added to the cultures at a final concentration range of 300nM to 10 nM. IvIG was used at a final concentration of 10. mu.g/mL. The cells were mixed by pipetting up and down and incubated at 37 ℃ for 18 hours. At the end of the incubation period, the cells were centrifuged at 400g for 5 minutes and resuspended in PBS. Thereafter, cells were stained with 1 μm Zombie viability dye (Biolegged, Cat. No. 423111) according to the manufacturer's instructions and in the chamber Incubate at room temperature for 15 minutes. The fluorescent antibody mixture was then added to cells containing CD56 PE, CD38 PE-Cy7, CD3 BV786, and CD107a BV421 in PBS 1% BSA, 0.5mM EDTA buffer. The cells were further incubated at 4 ℃ for 15 minutes and washed twice with 200. mu.L PBS 1% BSA, 0.5mM EDTA buffer. The cells were resuspended in a final volume of 150. mu.L PBS 1% BSA, 0.5mM EDTA buffer and 20. mu.L of Count BrightTMAbsolute count beads (seimer feishier catalog No. C36950) to determine absolute cell counts. Cells were analyzed on a BD FACSCelesta flow cytometer (BD Biosciences). NK cells are defined as CD3-CD56+. The percentage of Zombie green positive NK cells was analyzed and plotted using GraphPad Prism software. Data from one example is presented in fig. 4. As shown, the provided ARM did not deplete effector cells, e.g., NK cells, expressing CD 38. Various additional analyses, including in vivo animal models, demonstrate that the provided techniques can effectively kill diseased cells (e.g., cancer cells), do not induce NK cell suicide or CDC, and/or do not significantly affect immune cell populations (e.g., various types of immune cells, such as in bone marrow immune cell populations) as compared to CD38 antibodies (e.g., daratumab), demonstrate that the provided techniques can be highly efficient and do not cause significant side effects/toxicity as compared to antibody-based techniques.
Example 6. the provided technology can effectively kill cancer cells without causing significant side effects.
Cancer therapy can have various side effects. For example, therapy with antibodies against certain cancer antigens may harm, inhibit, or kill non-cancer cells that include the same antigen. Furthermore, the present disclosure presents the following results, demonstrating that the provided technology is effective against cancer cells expressing various antigens while having much less side effects/toxicity compared to other technologies (including antibody-based technologies) targeting the same antigen. For example, the provided techniques do not significantly reduce the number of non-cancerous cells expressing the same target as compared to other techniques.
Some applicable procedures
PBMC separation: from the bodyThe leukopheresis products of donors with a quality index (BMI) between 19 and 25, an age of less than 50 years, no immunosuppressive drugs administered for at least 2 weeks, an estimated PBMC cell count > 10^10 were obtained from KeyBiologics (goal ID 20982200) and shipped at ambient temperature. The leukopheresis bag was wiped with 70% EtOH, a small slit was cut, and the contents were pipetted into the test tube. The contents of Leukopak were diluted 1: 1 with PBS without Ca + +/Mg + +, and 20mL of this mixture was layered on top of 25mL Ficoll. The tube was then centrifuged at 400g for 30 minutes without interruption. PBMCs were collected from the interface into separate tubes, and the tubes were filled to 50mL with PBS. Cells were centrifuged at 120g for 10 min to remove platelets. The supernatant was decanted and the cells were resuspended in RPMI 10% FBS at a concentration of 7 to 10X 10^6 cells/ml. Cells were incubated at 37 ℃ with 5% CO 2Incubate under atmosphere overnight until NK cells detached.
And (3) NK cell separation: PBMC were centrifuged at 400g for 10 min and resuspended in EasySep buffer. NK cells were isolated using NK cell isolation kit from Stem cell technology, Inc according to the kit manufacturer's instructions. NK cell purity and phenotype were assessed by trypan Blue (trypan Blue) and flow cytometry using the following antibody cocktail: anti-CD 56PE, CD3 APC, and viability was determined by staining with trypan blue exclusion dye and by flow cytometry using near-infrared fixable viability dye (seemer feishol).
CIML NK cell production: isolated NK cells were resuspended in XVIVO 10% human serum and IL-15(50ng/ml), IL-12(10ng/ml), IL-18(50ng/ml) at 2X 10^6 cells/ml for 12 to 18 hours. Cells were collected and tested for purity and viability by trypan blue staining and by flow cytometry using near infrared fixable viability dyes (seemer feishel), CD3 APC (bio-legend) and CD56PE (bio-legend).
Treatment with I-17: i-17 was dissolved in DMSO to prepare a 25mM stock solution. The stock was then serially diluted in 100% DMSO to obtain the 1000 × concentration to be used for the experiment. Then 100% DMSO compound stock was added to the tube and the medium was added to a volume of 1000 μ Ι per 1 μ Ι compound and vortexed for 1 minute to ensure a homogeneous mixture. NK cell pellets were resuspended directly in I-17 solution at a cell density of 5X 10^6 cells/ml.
CIML NK cell self phase killing assay: NK cells were resuspended directly in the solutions generated for each concentration of I-17 at a density of 5X 10^6 cells/ml. Similarly, NK cell pellets were resuspended in Darandian single anti-control antibody solution (range of 3. mu.g/ml to 0.01. mu.g/ml in PBS 5% HSA). The resulting NK cell suspension was aliquoted at 55. mu.l per well of 96-well v-bottom plate and at 37 ℃ in 5% CO2Incubate under atmosphere for 2 hours.
CIML NK cells SUDHL-4 cell ADCC assay: SuDHL-4 cells were labeled with CFSE and resuspended in XVIVO15 medium containing 20% human serum. Cells were aliquoted at 10,000 cells per well, 18 μ Ι per well in a 96 well V-bottom plate. NK cells (in PBS 5% HSA) were added in equal volumes (18. mu.l per well) at a 9: 1 ratio (90,000 NK cells per well). The CO-culture was incubated at 37 ℃ in 5% CO2Incubate under atmosphere overnight. The following day, cells were washed and stained with the following reagents: near infrared fixable vital dyes (seimenfiel), CD3 FITC (bioglass) and CD56 PE (bioglass). The percentage of dead SUDHL-4 cells was calculated by gating on CFSE + cells that were also positive for near-infrared vital stain.
Some results
Purity and viability of NK cells and CIML NK cells: in addition, the present disclosure provides a population of NK cells with high purity and viability following isolation from PBMCs. Peripheral blood mononuclear cells were isolated to form leukodepleted products, with > 99% PBMC viability after isolation as determined by trypan blue and by flow cytometric analysis. After separation using magnetic bead assisted negative selection, NK cells were 90% pure and more than 99% viable as determined by both trypan blue and flow cytometry. T cell contamination (CD3+ CD 56-cells) in the NK cell fraction was found to be 2.8% on the day of isolation. After isolation of NK cells, they were divided into two treatment groups, CIML NK cells and non-CIML (control cells). CIML NK cells were incubated overnight in XIVO15 medium containing 10% human serum and IL-12, IL-15 and IL-18, while control non-CIML NK cells received no cytokine treatment. After overnight incubation, purity and viability were assessed in the cultured cell populations. In one experiment, the viability of CIML and non-CIML control NK cells determined by trypan blue was 50%. The viability of CIML NK cells determined by flow cytometry was 75%, whereas the viability of control non-CIML NK cells determined by flow cytometry method was 78%. At the time of harvest, T cell contamination in both cultures ranged from 1.2% to 1.8%.
The provided technology exhibits significantly lower toxicity compared to a corresponding technology comprising antibodies directed against the same target.
NK cell suicide (NK cell-directed death in the same culture at NK cells) was assessed by flow cytometry after 2 hours of incubation with the indicated concentration of I-17 in the presence or absence of 500 μ g/ml intravenous immunoglobulin (IVIG). Figure 5 shows the frequency of dead NK cells as a function of I-17 concentration in daratumab, CD 38-directed therapeutic antibody, or culture supernatant.
As shown, the provided techniques exhibit significantly lower toxicity compared to corresponding techniques using antibodies, e.g., when assessed by a reduction in cell number of non-cancer cells. For example, at only 2 hours after incubation, 3 μ g/ml or 20.1nM of daptomu mono-antibody treatment resulted in a 4% increase in the percentage of dead NK cells present in the cultured medium compared to no increase in the case of I-17+/-IVIG for CIML NK cells (FIG. 5). In cultures of non-CIML NK cells, treatment with daptomycin produces a total increase of 5.6% dead NK cells compared to DMSO-treated controls, whereas treatment with the highest concentration of I-17(25 μ M) caused a 1.7% increase (25 μ M of I-17 versus DMSO controls) and a 0.93% increase (I-17 IvIG versus DMSO IVIG controls) in dead NK cells. Figure 5(B) presents data normalized to DMSO-treated controls. As shown, no significant NK cell suicide was detected even at the highest dose of I-17, in sharp contrast to darunavir (a CD38 therapeutic antibody approved by the FDA for the treatment of multiple myeloma).
The provided technology can effectively reduce the number of target cancer cells.
Antibody-dependent cytotoxicity assays were performed to measure the effect of CIML NK cells alone and in combination with I-17 on SUDHL-4 multiple myeloma cells of interest. Baseline activity was assessed by using NK cells that had not been stimulated with cytokine cocktail (non-CIML NK cells). FIG. 6 presents the frequency of dead SUDHL-4 cells in NK-SUDHL-4 cell co-cultures after 18 hours of incubation.
As shown from the data presented in fig. 6B, CIML NK cells are potent killers of SUDHL-4 target cells compared to baseline (DMSO-treated control). Additional target cell killing compared to DMSO-treated control levels can be achieved by CIML NK cells in combination with I-17-an increase in killing of 66% compared to background was observed. Furthermore, as shown herein, the provided techniques can effectively utilize IgG present in human serum, and do not require the administration of additional antibodies, such as exogenous antibodies similar to those used in antibody-based therapies. Data using darunavir targeting CD38 is also included. As demonstrated herein, the provided techniques kill cancer cells efficiently while being less toxic than antibody-based techniques.
Example 7. the provided technology can effectively kill a given cell with low toxicity.
Furthermore, this example demonstrates that the provided techniques can effectively reduce the number of cancer cells. In this example, I-17 (as in the examples above), cytokine-induced memory-like NK cells (CIML NK cells) and optionally intravenous immunoglobulin (IVIG) were utilized. In addition, the present disclosure demonstrates the activity of the provided technology against bone marrow plasma cells in multiple myeloma patients.
Materials, methods and apparatus
Test and control articles
I-17 (25 mM solution in DMSO)
Darlazeux (Darzalex) -Parexel DRE0607
Intravenous IVIG human immunoglobulin, Flebogama-Grifols NDC 61935-
Materials and apparatus
Bovine Serum Albumin (BSA) heat shock, fraction V-American organism AB01088-00100
Phosphate Buffered Saline (PBS) -Gibco 10010, (-) Ca (-) Mg, pH 7.4
Intravenous IVIG human immunoglobulin, Flebogama-Grifols NDC 61935-
biotinylated/His-tagged human CD38 protein, Avi Tag-Popseris (Acro Biosystems) CD8-H82E7
Neutravidin-Invitrogen (31000, 1 mg/ml)
Dimethyl sulfoxide (DMSO), Sigma-Aldrich 276855
Bovine Serum Albumin (BSA) heat shock, fraction V-American organism AB01088-00100
Ethylenediaminetetraacetic acid (EDTA) VWR 97062-656
Running buffer-PBS pH 7.4+ 0.05% (w/v) BSA, 2mM EDTA
Assay plate, 96-well V-bottom-VWR corning 3357
Attune NxT flow cytometer and Attune NxT software
96 well 2ml deep well plate, catalogue number 1896-
Ficoll (R) Paque Plus, GE Healthcare group catalog number 17-1440-03
EasySEPTMHuman NK cell isolation kit, catalog No. 17955 of Stem cell technology Co., Ltd
Premium grade human IL-15, Cat. Ataland Biotech (Miltenyi Biotec) catalog number 130-
Premium grade human IL-12, catalog number 130-
Human IL-18, Andi Biopsis (R & D Systems) catalog number 9124-IL-010
Special cell culture medium, Lonza X-VIVO15, Saimer Feishell scientific catalog number BW04-744Q
Human AB blood clot serum, Gemini Bioproducts catalog number 100-
Albumin from human serum, Sigma-Aldrich catalog A5843-5G
RBC lysis buffer 10X (Tonbo Biosciences catalog number TNB-4300-L100)
Minimal residual disease antibody staining group:
Antigens Fluorophores Cloning Suppliers of goods Directory number
CD138 BV421 MI15 Biological science of BD 562935
CD27 BV510 O323 Biographical legends 302835
CD38ME FITC Multi-epitope Cytognos CYT-38F2
CD56 PE 5.1H111 Biographical legends 362508
CD45 PERCPCY5.5 HI30 Biographical legends 304028
CD19 PECY7 HIB19 Biographical legends 302216
CD117 APC 104D2 BD 341096
Patient bone marrow and blood were obtained from Discovery Life Sciences (Discovery Life Sciences).
Scheme(s)
Bone marrow (3ml) and blood (20ml) from patients found in Biosciences (Discovery Biosciences) were transported at ambient temperature. Bone marrow was diluted 1: 1 with PBS and 50^ l of bone marrow-PBS mixture was aliquoted into 96-deep well plates. Bone marrow cells were incubated overnight until the next day CIML cells were added.
PBMCs were isolated from blood by Ficoll density gradient centrifugation. PBMC were centrifuged at 400g for 10 min and resuspended in EasySep buffer. NK cells were isolated using NK cell isolation kit from Stem cell technology, Inc according to the kit manufacturer's instructions.
NK cells were cultured at 1X 106Each cell/ml was resuspended in XVIVO15, 10% human serum, IL-15(50ng/ml), IL-12(10ng/ml), IL-18(50ng/ml) and incubated for 18 hours. After overnight incubation with cytokines, NK cells were washed twice with PBS 5% human serum albumin, counted and cultured at 2 × 10 per ml XVIVO15 medium 5Cell density of individual cells was resuspended. To a cell containing 50^ l bone marrow: twenty microliters (4000 cells) was added to each well of the PBS mixture. Mixing dacematoclone antibody (3^ g/ml [20 nM)]To 0.3^ g/ml [2nM]) And I-17(25^ M to 3^ M final concentration) was made 10X of the final concentration to be used for analysis, and 8^ l of I-17 or 7^ l of Darasba monocarb 10X stock solution was added to the relevant well. To I-17 treated wells, human IVIG was added to a final concentration of 10^ g/ml. Control wells received XVIVO15 medium. The well contents were mixed by pipetting up and down and the plate was centrifuged at 400g for 1 minute.
Then at 37^ C, 5% CO2The lower incubation was analyzed for 4 hours. After incubation, 1ml of 1 × erythrocyte lysis buffer was added to each well and the cells were incubated at room temperature for 15 minutes. Plates were centrifuged at 400g for 3 minutes and another round of erythrocyte lysis and centrifugation was performed. The cells were then resuspended in 200^1 FACS buffer and centrifuged. The buffer was discarded and the cells were resuspended in 100^1 fluorescent antibody mixture. The cells were incubated at 4^ C for 15 minutes, 200^1 FACS buffer was added to each well for washing, the cells were centrifuged at 400g for 3 minutes, and the supernatant was discarded. The washing was repeated 2 more times. Finally, the cells were resuspended in 200^1 FACS buffer and analyzed using an Attune flow cytometer. Data were analyzed using FlowJo software and graphed using GraphPad Prism.
The provided techniques reduce the number of plasma cells.
FIG. 7 shows the percentage reduction in CD38+ CD138+ cell frequency in the bone marrow of patients after 4 hours of incubation in the presence of autologous CIML NK cells and daratumab or I-17. As shown in FIG. 7, I-17 effectively reduced the frequency of plasma cells (CD38+ CD138+) compared to control untreated bone marrow incubated with CIML NK cells.
The provided techniques do not significantly affect the immune cell population.
Furthermore, this example further demonstrates that the provided technology has low toxicity, especially when compared to therapies where the antibodies utilized are directed to the same target as ARM. As shown in FIG. 8 (frequency of CD56+ CD138-CIML NK cells when co-incubated with patient bone marrow and either daratumab or I-17 for 4 hours), NK cell number did not change in the presence of I-17, whereas darunavir showed a dose-dependent increase in NK cell suicide.
Example 8. the provided techniques can effectively reduce the number of target cells.
As described herein, the provided techniques can effectively reduce the number of target cells (e.g., cells expressing CD 38). Without intending to be limited by theory, in some embodiments, the reduction in the number of target cells may include or be mediated by ADCP, which in some cases is mediated by macrophages. Techniques for assessing provided compounds, compositions, and methods, as well as illustrative data demonstrating the activity of provided techniques (e.g., compounds, compositions, methods, etc.) are described below.
Materials and methods
Test system
Species/strain: mouse/SCID
Physiological state: is normal
Age/body weight range at study start: the animals were about 6 weeks old and about 25g in weight.
Animal suppliers: charles River Laboratory (Charles River Laboratory)
Number/sex of animals 32/female
Marking: ear number/transponder
Randomization: animals were randomized prior to assignment to treatment groups.
Adjusting: mice represent a well characterized system for drug efficacy assessment.
And (3) replacing: animals will not be replaced during the course of the study.
Design of experiments
On day 0, animals were weighed and randomized. Mice were assigned to different groups and treated as described below. CFSE labeled Daudi cells were prepared as follows:
CFSE staining of Daudi cells for IP injection:
the reagents used were: PBS (sterile)
Quenching buffer PBS + 10% FBS
RPMI (no additive), no P/S; FBS-free
Invitrogen-CellTrace CFSE cell proliferation kit for flow cytometry (Cat. No. C34570)
Sterile PBS and all other media were warmed to 37 degrees celsius prior to use for all the following steps listed below.
Sufficient cells were collected from the cell culture to contain 50% more cells than required for the analysis
Cells were washed 2 x with sterile PBS by centrifugation at 500g for 7 min.
The cells were resuspended in sterile PBS in a 50mL polypropylene tube to prepare a cell suspension such that the cell suspension occupied less than 1/4 of the total tube volume.
A 5mM stock solution of CFSE was prepared by adding 18 μ L DMSO to a lyophilized tube of CFSE.
The solution was diluted 10-fold from the stock solution in PBS to make a 100X (500. mu.M) solution
Add 10. mu.L or 100 XCFSE solution per 1mL of cells
CFSE stock solution was added to the resuspended cells so that the final concentration of CFSE was 5 μ M.
The cell suspension was gently mixed by vortexing until the density stabilized dmso (cfse) was no longer visible.
The cap of the 50mL conical tube (containing CFSE-labeled cells) was released, it was stood in a test tube rack, and it was placed in CO at 37 deg.C2The incubator was left for 10 minutes while vortexing every 2 minutes.
After incubation, the tube was topped up with quench buffer for quenching and allowed to stand for 2 minutes.
Cells were washed 2 x by centrifugation at 500g for 7 min and rewashed with sterile quench buffer.
Cells were washed 3 x with cold sterile RPMI (without additives) by centrifugation at 500g for 7 min.
For each washing step, the volume is at most 50mL
Final cell count was performed:
cells were diluted 1: 50 in counting vials
Resuspend to desired cell density with approximate volume of cold RPMI (no additive):
modulating cell density
Cells will be injected per mouse: 400 μ L per mouse; total 20X 10 per mouse6An
After 16 to 18 hours, via CO2Mice were sacrificed by asphyxiation and their peritoneal cavity was rinsed with 5ml PBS containing 1% BSA. The samples were processed and analyzed using the IP lavage process described in 00765. The samples were evaluated for tumor cell counts.
Figure BDA0003526174380003881
Study schedule:
Figure BDA0003526174380003882
animal body weights were measured and recorded at day 0 randomization. Additional body weight was collected at the time of administration of the same compound.
And (4) collecting tissues. After 16 to 18 hours, via CO2Mice were sacrificed by asphyxiation and their peritoneal cavity was rinsed with 5mL PBS containing 1% BSA. The samples were kept on ice until further processing.
Applicable IP lavage treatment protocol:
1. intraperitoneal lavage was collected using 5ml of IgG free PBS 1% BSA.
2. Lavage samples were briefly centrifuged at 400g for 5 minutes.
3. The maintenance marks the tube as originally received from the animal facility and keeps the sample on ice.
4. The volume of each sample was recorded on a spreadsheet.
5. The supernatant was poured into 5ml Eppendorf (Eppendorf) tubes and stored at-80.
6. The pellet was resuspended in 250 μ L of PBS 1% BSA without IgG and transferred to a blue top filter flow tube.
7. The top filter tube was centrifuged briefly at 400g for 5 minutes.
8. The supernatant was poured into a waste tube.
RBC present? The following steps are continued:
a. RBC lysis was performed by resuspending the pellet in 1ml of RBC lysis buffer for 5 minutes at room temperature, adding 1ml of PBS 1% BSA buffer to each tube, and centrifuging for 5 minutes at 400g briefly.
b. The pellet was resuspended in 250 μ L of IgG-free PBS 1% BSA by pipetting up and down, and the whole cell suspension was transferred to a 96-well plate labeled with the sample number.
c. Plates were centrifuged briefly at 400g for 3 minutes and the supernatant was flicked off, and plates were placed on ice.
Applicable preparation protocol for flow analysis on Attune:
1. cells were resuspended in 250 μ L PBS 1% BSA without IgG in tubes.
2. Samples from the above steps were divided into 2 identical 96-well V-bottom plates.
3. And (3) fixing procedures:
a. fixation was performed in 4% paraformaldehyde (stock 16%, diluted 1: 4 with PBS).
b. Plates were centrifuged briefly at 400g for 5 minutes and the supernatant was flicked off.
c. Resuspend with 125 μ l IgG free PBS 1% BSA.
d. Add 125. mu.l of 4% paraformaldehyde with a pipette and mix gently.
e. Incubate at room temperature for 10 minutes.
f. Plates were centrifuged briefly at 400g for 5 minutes and the supernatant was flicked off.
g. Resuspended in 250. mu.l PBS 1% BSA without IgG.
h. Stored at 4 degrees celsius.
4. Run fixed samples:
a. the plate is removed from the refrigerator.
b. Plates were centrifuged briefly at 400g for 5 minutes and the supernatant was flicked off.
c. The CST beads were run on Attune and the experiment set up while the plate was briefly centrifuged.
d. Resuspend with 250 μ l of IgG free PBS 1% BSA.
Some of the data is presented in fig. 9. As demonstrated herein, the provided techniques can effectively reduce the number of target cells, as demonstrated by the dose-dependent reduction of Daudi cells in an intraperitoneal lavage model, which can be mediated via or including macrophage-dependent ADCP of Daudi cells, without intending to be limited by theory.
Example 9 compositions comprising cells can be cryopreserved.
In addition, the present disclosure demonstrates that compositions including cells can be cryopreserved and later utilized to provide a desired activity. In the examples:
CIML NK cells were produced from the same day arriving white blood cell package (leukopack) and had greater than 95% viability prior to freezing.
PBMC, NK cell isolation and CIML production were as described (e.g., example 6; scientific transformed Medicine, Romee et al, 2016, 9, 21, 9, 8, 357ra 123; digital object identifier: 10.1126/scitranslim. aaf2341).
3. CIML NK cells were washed free of cytokines with RPMI 10% FBS and twice.
4. Viability was assessed by trypan blue or L/D identification dyes (e.g., L/D near IR) prior to freezing.
5. Cells were frozen in FBS 10% DMSO at 100X 10^6 cells per vial per ml, or in the same medium + 25. mu. M I-17:
briefly, a 25mM stock of I-17 was generated in DMSO.
This was further diluted 1: 100 in DMSO to make 250. mu.M.
Preparing a freezing culture medium:
Figure BDA0003526174380003901
does not contain I-17: 90% FBS + 10% DMSO-was used for control cells.
Figure BDA0003526174380003902
Contains I-17: 90% FBS + 10% in DMSO 250 u M I-17 solution-for and I-17 together with frozen cells.
6. Cells were placed in a mister frost isopropanol container and stored at 80 ℃.
7. Cells were maintained at-80 for 1 day and then switched to liquid nitrogen.
8. Cells were maintained in liquid nitrogen for 2 weeks before thawing.
9. The vial was placed at 37H2Thaw in O bath for more than 5 minutes.
10. Added dropwise to 35mL of warm RPMI 10% FBS.
11. Centrifuge at 400g for 10 min.
12. Wash 1 x with RPMI 10% FBS.
13. Centrifuge at 400g for 10 min.
14. The viability is counted and assessed using a near IR viability dye or the like.
15. Proceeding to ADCC: the CFSE-labeled MOLP8 cells were resuspended in 10 k/50. mu.l RPMI 10% FCS.
16. Add to round bottom 96-well plates.
17. NK cells were resuspended at 100K/50. mu.L, 50K and 25K and added to the target cells.
18. To CIML cell conditions were added 3. mu.g/mL of daratumab or 2.5. mu.M of I-17+ 10. mu.g/mL of IvIG.
19. To the wells receiving CIML NK cells frozen in combination with I-17, only IvIG was added.
20. Incubate for 4 hours.
21. Staining was done with IR L/D, CD107a and CD 69.
As shown in fig. 10, a composition comprising cells can be cryopreserved and then thawed and used with provided compounds (provided compounds in the same cryopreserved composition and/or added separately) to provide activity. In some embodiments, the cells are stored with a provided compound (e.g., an ARM agent). In some embodiments, the cells are not stored with the ARM agent provided, which can be added separately.
While we have described a number of embodiments, it is apparent that our basic examples can be modified to provide other embodiments that utilize the compounds and methods of the present disclosure. It is, therefore, to be understood that the scope of the invention should be defined by the appended claims rather than the specific embodiments illustrated.

Claims (72)

1. A medicament, comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
the optional presence of a linker moiety or moieties,
wherein the target binding moiety specifically binds to CD 38.
2. The agent of claim 1, wherein the agent has the structure of formula I:
Figure FDA0003526174370000011
each of a and b is independently 1 to 200;
each ABT is independently an antibody binding moiety;
l is a linker moiety connecting ABT and TBT; and is
Each TBT is independently a target binding moiety.
3. The agent of claim 1, wherein the agent has the structure:
Figure FDA0003526174370000012
or a pharmaceutically acceptable salt thereof, wherein:
and b is independently from 1 to 200;
each ABT is independently an antibody binding moiety;
l is a bivalent linker moiety linking ABT to TBT;
each Xaa is independently a residue of an amino acid or amino acid analog;
y is 5 to 20;
LTTo link two of amino acids or amino acid analoguesLinker moieties of each independent residue, and independently being a covalent bond, or selected from C1-C6Aliphatic radical or C having 1 to 5 hetero atoms1-C6A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each RcIndependently is-La-R′;
t is 0 to 50;
each LaIndependently a covalent bond, or is selected from C1-C50Aliphatic radical or C having 1 to 5 hetero atoms1-C50A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from C3-20Cycloaliphatic Ring, C6-20An aryl ring, a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms, and a 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30Aliphatic radical, C having 1 to 10 heteroatoms 1-30Heteroaliphatic radical, C6-30Aryl radical, C6-30Arylaliphatic radical, C having 1 to 10 heteroatoms6-30Aryl heteroaliphatic, 5-to 30-membered heteroaryl having 1 to 10 heteroatoms and 3-to 30-membered heterocyclyl having 1 to 10 heteroatoms, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, together with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the atom; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the intervening atoms.
4. A medicament, comprising:
(ii) an antibody-binding moiety,
a target binding moiety, and
the optional presence of a linker moiety or moieties,
wherein the target binding moiety has the following structure:
Figure FDA0003526174370000031
each Xaa is independently a residue of an amino acid or amino acid analog;
y is 5 to 20;
LTa linker moiety which is two separate residues of an amino acid or amino acid analogue and is independently a covalent bond, or is selected from C 1-C6Aliphatic radical or C having 1 to 5 hetero atoms1-C6A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each RcIndependently is-La-R′;
t is 0 to 50;
each LaIndependently a covalent bond, or is selected from C1-C50Aliphatic radical or C having 1 to 5 hetero atoms1-C50A heteroaliphatic optionally substituted divalent radical wherein one or more methylene units of said radical are optionally and independently replaced by-C (R')2-、-Cy-、-O-、-S-、-S-S-、-N(R′)-、-C(O)-、-C(S)-、-C(NR′)-、-C(O)N(R′)-、-N(R′)C(O)N(R′)-、-N(R′)C(O)O-、-S(O)-、-S(O)2-、-S(O)2N (R') -, -C (O) S-or-C (O) O-substitution;
each-Cy-is independently an optionally substituted divalent monocyclic, bicyclic, or polycyclic group, wherein each monocyclic ring is independently selected from C3-20Cycloaliphatic Ring, C6-20An aryl ring, a 5-to 20-membered heteroaryl ring having 1 to 10 heteroatoms, and a 3-to 20-membered heterocyclyl ring having 1 to 10 heteroatoms;
each R' is independently-R, -C (O) R, -CO2R or-SO2R;
Each R is independently-H, or an optionally substituted group selected from: c1-30Aliphatic radical, C having 1 to 10 heteroatoms1-30Heteroaliphatic radical, C6-30Aryl radical, C6-30Arylaliphatic radical, C having 1 to 10 heteroatoms6-30Aryl heteroaliphatic, 5-to 30-membered heteroaryl having 1 to 10 heteroatoms and 3-to 30-membered heterocyclyl having 1 to 10 heteroatoms, or
Optionally and independently, two R groups together form a covalent bond, or:
two or more R groups on the same atom optionally and independently form, together with the atom, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the atom; or
Two or more R groups on two or more atoms optionally and independently form, together with their intervening atoms, an optionally substituted 3-to 30-membered monocyclic, bicyclic, or polycyclic ring having 0 to 10 heteroatoms in addition to the intervening atoms.
5. The medicament of claim 4, wherein- (Xaa) y-comprises:
-XaaT1-XaaT2-(Xaa)y′-XaaT3-XaaT4-XaaT5-,
wherein:
y' is 0 to 8;
XaaT1for C whose side chain is substituted1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT2including an optionally substituted aromatic group for its side chain or being optionally substituted C3-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT3is C whose side chain is optionally substituted2-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT4including an optionally substituted aromatic group for its side chain or being optionally substituted C3-C8A residue of an amino acid or amino acid analog of an aliphatic group; and is
XaaT5For C whose side chain is substituted 1-C8Aliphatic amino acids or amino acid analogues.
6. The medicament of claim 5, wherein XaaT1Is the residue of Ahp, Y, W, S, K or K (MePEG4 c).
7. The medicament of claim 6, wherein XaaT2Is the residue of Y, W, Ahp, Bph, L or A.
8. The medicament of claim 7, wherein XaaT3Is a residue of L, Ahp, V, T, Hse or MetO 2.
9. The medicament of claim 8, wherein XaaT4Residues of Bph, V or Ahp.
10. The medicament of claim 9, wherein XaaT5Residues of Ahp, Bph, Ado, Ano, PhNle or PhNva.
11. The medicament of claim 10, wherein- (Xaa) y-is or comprises:
-(Xaa)a1-(Xaa)a2-(Xaa)a3-(Xaa)a4-(Xaa)a5-(Xaa)a6-(Xaa)a7-(Xaa)a8-(Xaa)a9-(Xaa)a10-(Xaa)a11-(Xaa)a12-(Xaa)a13-,
wherein:
each of a1, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, a12, and a13 is independently 0 to 5;
(Xaa)a3is or comprises XaaT1
(Xaa)a4Is or comprises XaaT2
(Xaa)a9Is or comprises XaaT3
(Xaa)a10Is or comprises XaaT4(ii) a And is
(Xaa)a11Is or comprises XaaT5
12. The medicament of claim 11, wherein (Xaa)a1Is or includes A, K or K (MePEG4 c).
13. The medicament of claim 12, wherein (Xaa)a2Is or includes R, S, D, Y, A, W, K, 4Py2NH2, Cit, F3G, hCit, K (MePEG4c), RNdMe, RNMe or RNNdMe.
14. The medicament of claim 13, wherein (Xaa)a5Is or includes H, Y, S, L, A, W or W6N.
15. The medicament of claim 14, wherein (Xaa)a6Is or includes D, G, R, Y, H, W, A or Y.
16. The method of claim 15Medicament, wherein (Xaa)a7Is or includes G, D, E, Q, N, R, MetO2, S, Har or A.
17. The medicament of claim 16, wherein (Xaa)a8Is or includes V, A, D, G, W, S or T.
18. The medicament of claim 17, wherein (Xaa)a12Is or includes D, A, S, G or Ahp.
19. The medicament of claim 18, wherein (Xaa)a13Is or includes C.
20. The medicament of claim 4, wherein- (Xaa) y-comprises:
-XaaT6-(Xaa)y′-XaaT7-XaaT8-XaaT9-XaaT10-XaaT11-,
wherein:
y' is 0 to 8;
XaaT6for C whose side chain is substituted1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT7is C whose side chain is optionally substituted2-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT8is a residue of proline or an amino acid analogue thereof;
XaaT9including an optionally substituted aromatic group for its side chain or being optionally substituted C1-C8A residue of an amino acid or amino acid analog of an aliphatic group;
XaaT10for C whose side chain is substituted 1-C8A residue of an aliphatic group amino acid or an amino acid analog or a residue of an amino acid in which an amino group thereof is substituted; and is
XaaT11Including an optionally substituted aromatic group for its side chain or being optionally substituted C1-C8Aliphatic amino acidsOr a residue of an amino acid analog.
21. The medicament of claim 20, wherein XaaT6Is a residue of MeF, L or S.
22. The medicament of claim 21, wherein XaaT7Is a residue of L or MeF.
23. The medicament of claim 22, wherein XaaT8Is the residue of P.
24. The medicament of claim 23, wherein XaaT9Is a residue of Bph, D or S.
25. The medicament of claim 24, wherein XaaT10Is a residue of V or L.
26. The medicament of claim 25, wherein XaaT11Is a residue of W or R.
27. The medicament of claim 26, wherein- (Xaa) y-is or comprises:
-(Xaa)a1-(Xaa)a2-(Xaa)a3-(Xaa)a4-(Xaa)a5-(Xaa)a6-(Xaa)a7-(Xaa)a8-(Xaa)a9-(Xaa)a10-(Xaa)a11-(Xaa)a12-,
wherein:
each of al, a2, a3, a4, a5, a6, a7, a8, a9, a10, a11, and a12 is independently 0 to 5;
(Xaa)a4is or comprises XaaT6
(Xaa)a6Is or comprises XaaT7
(Xaa)a7Is or comprises XaaT8
(Xaa)a8Is or comprises XaaT9
(Xaa)a9Is or a bagInclude XaaT10(ii) a And is
(Xaa)a10Is or comprises XaaT11
28. The medicament of claim 27, wherein (Xaa)a1Is or includes A.
29. The medicament of claim 28, wherein (Xaa)a2Is or includes L, A or P.
30. The medicament of claim 29, wherein (Xaa)a3Is or includes H, R or A.
31. The medicament of claim 30, wherein (Xaa)a5Is or includes V, A or MeG.
32. The medicament of claim 31, wherein (Xaa)a11Is or includes V, A, D or MeG.
33. The medicament of claim 32, wherein (Xaa)a12Is or includes C.
34. The agent of claim 4, wherein the target binding moiety or
Figure FDA0003526174370000061
Is or comprises a sequence selected from SEQ ID NO: 1-34.
35. The medicament of any one of claims 4 to 34, wherein two Xaa are linked together.
36. The medicament of claim 35, wherein two Xaa are via a c- (o) -CH having2-the linkers of the structure are linked together.
37. The agent of claim 36, wherein-c (o) -amino bonded to Xaa.
38. The agent of claim 37, wherein-CH2-S-bonded to the side chain of Xaa.
39. The agent of claim 4, wherein the target binding moiety or
Figure FDA0003526174370000062
Is or comprise
Figure FDA0003526174370000071
Figure FDA0003526174370000081
Figure FDA0003526174370000082
Or a salt form thereof.
40. The agent of any one of claims 4-34, wherein the antibody binding portion can bind to two or more antibodies having different Fab regions.
41. The agent of any one of claims 4-34, wherein the antibody-binding portion has the structure DCAWHLGELVWCT or a salt form thereof, wherein two C residues are linked by-S-.
42. The agent of any one of claims 4-34, wherein the antibody binding moiety is or comprises optionally substituted
Figure FDA0003526174370000083
Figure FDA0003526174370000084
43. The agent of any one of claims 4 to 34, wherein the antibody binding moiety is or comprises
Figure FDA0003526174370000091
44. The agent of any one of the preceding claims, comprising a linker.
45. The agent of any one of the preceding claims, wherein the linker is or comprises- (CH)2CH2O) n-where n is 1 to 20.
46. The agent of any one of the preceding claims, wherein the linker comprises one or more amino acid residues.
47. The agent of any one of the preceding claims, wherein the linker comprises
Figure FDA0003526174370000092
48. A medicament, wherein the medicament is I-1, I-2, I-3, I-4, I-5, I-6, I-7, I-8, I-9, I-10, I-11, I-12, I-13, I-14, I-15, I-16, I-17, I-18, I-19, I-24, I-25, I-26, I-27, I-28, I-29, I-30, I-31, I-32, I-33, I-34, I-35, I-36, I-37, I-38, I-39, I-40, I-41, I-42, I-43, I-44, I-45, I-46 or I-47 or a pharmaceutically acceptable salt thereof.
49. The agent of any one of the preceding claims, wherein the agent binds to CD38 with a Kd of no more than 200, 100, 50, 40, 30, 20, 10, or 5nM as measured by SPR.
50. A composition comprising the agent of any one of the preceding claims and a population of cells.
51. The composition of claim 50, wherein the cell is or comprises an NK cell, an engineered NK cell, an ex vivo expanded NK cell, a memory-like NK cell, a cytokine-induced memory-like NK cell, an NKT cell, a monocyte and/or a macrophage.
52. A pharmaceutical composition comprising the agent or composition of any one of the preceding claims and a pharmaceutically acceptable carrier.
53. The composition of any one of claims 50-52, wherein the composition comprises an immunoglobulin.
54. The composition of claim 53, wherein the immunoglobulin is an intravenous immunoglobulin.
55. A method for treating a CD 38-associated condition, disorder or disease, comprising administering to a subject suffering from the CD 38-associated condition, disorder or disease an effective amount of the agent or composition of any one of the preceding claims.
56. The method of claim 55, comprising administering to the subject a population of cells.
57. The method of claim 56, wherein the subject is subjected to both the agent or composition and the population of cells.
58. The method of any one of claims 56-57, wherein the cell is or comprises an NK cell, an engineered NK cell, an ex vivo expanded NK cell, a memory-like NK cell, a cytokine-induced memory-like NK cell, an NKT cell, a monocyte and/or a macrophage.
59. The method of any one of claims 56-58, wherein the cell is administered concurrently with the agent or composition.
60. The method of any one of claims 56-59, wherein the cell and the agent or composition are administered simultaneously in the form of a composition comprising the cell and the agent or composition.
61. The method of any one of claims 56-58, wherein the cells are administered before or after administration of the agent or composition.
62. The method of any one of claims 55-61, wherein the method comprises administering intravenous immunoglobulin.
63. The method of claim 62, wherein the immunoglobulin is administered simultaneously with, before, or after the agent or composition.
64. The method of any one of the preceding claims, wherein one or more doses of the cells of any one of the preceding claims and/or one or more doses of the cells of any one of the preceding claims and the agent of any one of the preceding claims are administered after administration of the agent.
65. A method, comprising:
a) providing a first compound comprising a target binding moiety as described in any one of the preceding claims and a first reactive group;
b) providing a second compound comprising an antibody binding moiety as described in any one of the preceding claims and a second reactive group; and
c) reacting the first reactive group with the second reactive group to covalently link the target binding moiety to the antibody binding moiety.
66. The method of claim 65, wherein the first compound is a compound of formula V or a salt thereof.
67. The method of claim 65 or 66, wherein the second compound is a compound of formula IV, IV-a, IV-b, IV-c, or IV-d, or a salt thereof.
68. A method for manufacturing the agent or composition of any one of the preceding claims, comprising reacting a first compound comprising an antibody binding moiety and an alkyne with a second compound comprising a target binding moiety and an azide, or reacting a first compound comprising an antibody binding moiety and an azide with a second compound comprising a target binding moiety and an alkyne.
69. A method for recruiting an antibody to a target comprising or expressing CD38, comprising contacting the target with an agent or composition of any one of the preceding claims.
70. A method for recruiting immune activity to a target comprising or expressing CD38, comprising contacting the target with an agent of any one of the preceding claims.
71. The method of any one of claims 69-70, wherein the target is a tumor cell.
72. A compound, agent, composition or method as described in any one of embodiments 1-367.
CN202080061542.3A 2019-07-03 2020-06-24 CD38 binding agents and uses thereof Pending CN114401732A (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201962870633P 2019-07-03 2019-07-03
US62/870,633 2019-07-03
US201962951765P 2019-12-20 2019-12-20
US62/951,765 2019-12-20
PCT/US2020/039466 WO2021003050A2 (en) 2019-07-03 2020-06-24 Cd38-binding agents and uses thereof

Publications (1)

Publication Number Publication Date
CN114401732A true CN114401732A (en) 2022-04-26

Family

ID=74101217

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202080061542.3A Pending CN114401732A (en) 2019-07-03 2020-06-24 CD38 binding agents and uses thereof

Country Status (9)

Country Link
US (1) US20230028880A1 (en)
EP (1) EP3993818A4 (en)
JP (1) JP2022539258A (en)
KR (1) KR20220042130A (en)
CN (1) CN114401732A (en)
AU (1) AU2020299157A1 (en)
CA (1) CA3143513A1 (en)
IL (1) IL289531A (en)
WO (1) WO2021003050A2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113945723B (en) * 2021-10-28 2024-03-12 复旦大学附属中山医院 System for predicting risk of occurrence of immune checkpoint inhibitor treatment-related pneumonia, storage medium and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018222987A1 (en) * 2017-06-01 2018-12-06 Tarveda Therapeutics, Inc. Targeted constructs
WO2019002842A1 (en) * 2017-06-26 2019-01-03 Bicyclerd Limited Bicyclic peptide ligands with detectable moieties and uses thereof
WO2019023501A1 (en) * 2017-07-26 2019-01-31 Kleo Pharmaceuticals, Inc. Universal abt compounds and uses thereof

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10675352B2 (en) * 2014-02-14 2020-06-09 Centrose, Llc Extracellular targeted drug conjugates

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018222987A1 (en) * 2017-06-01 2018-12-06 Tarveda Therapeutics, Inc. Targeted constructs
WO2019002842A1 (en) * 2017-06-26 2019-01-03 Bicyclerd Limited Bicyclic peptide ligands with detectable moieties and uses thereof
WO2019023501A1 (en) * 2017-07-26 2019-01-31 Kleo Pharmaceuticals, Inc. Universal abt compounds and uses thereof

Also Published As

Publication number Publication date
WO2021003050A3 (en) 2021-02-11
EP3993818A2 (en) 2022-05-11
WO2021003050A2 (en) 2021-01-07
KR20220042130A (en) 2022-04-04
CA3143513A1 (en) 2021-01-07
JP2022539258A (en) 2022-09-07
AU2020299157A1 (en) 2022-01-20
US20230028880A1 (en) 2023-01-26
EP3993818A4 (en) 2023-10-11
IL289531A (en) 2022-03-01

Similar Documents

Publication Publication Date Title
US9284297B2 (en) Halofuginone analogs for inhibition of tRNA synthetases and uses thereof
JP2023103222A (en) Universal ABT compounds and uses thereof
US20180369230A1 (en) Targeted selection of patients for treatment with specific cortistatin derivatives
US20230028880A1 (en) Cd38-binding agents and uses thereof
US11807633B2 (en) Chromene derivatives as inhibitors of TCR-Nck interaction
EP4125893A1 (en) Technologies for preventing or treating infections
CN113454124A (en) Combination therapy of ARM and natural killer cells
WO2022170155A2 (en) Technologies for preventing or treating infections
TW202408538A (en) Stat degraders and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination